EP1292671B1 - Method for obtaining characterised muscle-derived cell populations and uses - Google Patents

Method for obtaining characterised muscle-derived cell populations and uses Download PDF

Info

Publication number
EP1292671B1
EP1292671B1 EP01943584A EP01943584A EP1292671B1 EP 1292671 B1 EP1292671 B1 EP 1292671B1 EP 01943584 A EP01943584 A EP 01943584A EP 01943584 A EP01943584 A EP 01943584A EP 1292671 B1 EP1292671 B1 EP 1292671B1
Authority
EP
European Patent Office
Prior art keywords
cells
cell
population
culture
treatment
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
EP01943584A
Other languages
German (de)
French (fr)
Other versions
EP1292671A1 (en
EP1292671B9 (en
Inventor
Jean-Thomas Vilquin
Jean-Pierre Marolleau
Jacques Tremblay
Isabelle Robert
Brigitte Ternaux
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Assistance Publique Hopitaux de Paris APHP
Institut National de la Sante et de la Recherche Medicale INSERM
Association Francaise Contre les Myopathies
Original Assignee
Assistance Publique Hopitaux de Paris APHP
Institut National de la Sante et de la Recherche Medicale INSERM
Association Francaise Contre les Myopathies
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Assistance Publique Hopitaux de Paris APHP, Institut National de la Sante et de la Recherche Medicale INSERM, Association Francaise Contre les Myopathies filed Critical Assistance Publique Hopitaux de Paris APHP
Publication of EP1292671A1 publication Critical patent/EP1292671A1/en
Application granted granted Critical
Publication of EP1292671B1 publication Critical patent/EP1292671B1/en
Priority to CY20101100077T priority Critical patent/CY1109745T1/en
Publication of EP1292671B9 publication Critical patent/EP1292671B9/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0658Skeletal muscle cells, e.g. myocytes, myotubes, myoblasts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)

Definitions

  • Cell therapy is a technique with high potential for the treatment of many pathologies.
  • the principle of cell therapy is based on the possibility of reconstituting damaged tissue or restoring lost or altered tissue function within a tissue, from specific cells cultured ex vivo and transplanted to the diseased tissue.
  • Another advantage of cell therapy is the possibility of using the transplanted cells as a delivery platform for a biologically active product, if necessary after genetic modification of the cells before transplantation. Many cell therapy trials are described from primary cultures of different cell types.
  • Parkinson's disease include neuronal cell transplants performed for the treatment of Huntington's disease (1) or Parkinson's disease (2), Langerhan's islet cell transplants performed for the treatment of diabetes (3), or Myoblastic cell transplants performed for the treatment of Duchenne muscular dystrophy (4, 5, 6, 7) or after genetic modification of the cells, for the treatment of dwarfism (8), hemophilia (9) and Parkinson's disease (10).
  • the regeneration of skeletal muscle is ensured by the satellite cells which are myogenic myonuclear cells situated under the basal lamina of the muscular fibers. Following an injury, these cells leave the quiescent state to enter an active proliferation phase and take the name of myoblasts. Subsequently, the myoblasts fuse to form the myotubes.
  • Myoblast transplantation trials have been conducted in humans to treat Duchenne muscular dystrophy and Becker muscular dystrophy (4, 6, 7, 11). Although the functional effect of transplants described in these studies remains limited, no side effects have been reported in terms of infection or carcinogenesis.
  • myoblastic cells in the treatment of heart disease, and in particular of post-ischemic heart failure has been considered.
  • myocardial tissues lack stem cells that can form cadiomyocytes and regenerate tissues.
  • the most radical treatment of post-ischemic heart failure remains at present, heart transplantation.
  • heart transplantation the scarcity of grafts limits this therapeutic use.
  • Transplantation of muscle tissue cells into cardiac muscle has therefore been considered as an alternative to cardiac transplantation.
  • Myoblast transplantation studies in infarcted myocardial tissue have been performed in rats, rabbits or dogs (12, 13, 14). The results of these studies have shown the feasibility and some functional efficacy of such transplants.
  • Fetal cardiomyocyte transplant assays in a model of iatrogenic heart failure in mice also showed some functional benefit.
  • the use of fetal cells poses, for the clinical application, many ethical, immunological and cellular supply problems.
  • the use of a population of myoblastic cells derived from muscle Skeletal is therefore a particularly interesting alternative for the preparation of cell therapy product for the treatment of post-ischemic heart failure, or even for the treatment of heart diseases of various origins.
  • muscle tissue contains other cell types. In particular, certain cells of muscular origin could also be used for the reconstitution of a hematopoietic potential (15, 16).
  • An in vitro study has also demonstrated the presence in human muscle of progenitors that can differentiate in the long term into cartilaginous or bone tissue (17). Examples of media allowing differentiation into adipose, cartilage or bone tissue are described for mesenchymal stem cells (22).
  • myoblastic cells In order to obtain a sufficiently dense and rich population, or even pure populations of myoblastic cells, it has been proposed to select myoblastic cells on the basis of the expression of specific markers. Thus, the selection of myoblastic cells can be done by cloning the cells and subsequently characterizing the clones obtained by cytofluorimetry and then selecting the skeletal muscle cells expressing the CD56 antigen (7).
  • a direct method of sorting myoblastic cells expressing the CD56 antigen by flow cytofluorometry and its advantages for obtaining a pure culture of myoblasts are also described in the state of the art (21).
  • the conserved cells are then cultured in a modified culture medium and specially adapted for culturing the myoblasts (23).
  • the present invention results from the observation that cells derived from skeletal muscle tissue have a potential for regeneration of many tissues depending on their degree of differentiation.
  • the proposed invention therefore makes it possible to provide well-characterized cell populations of muscular origin.
  • Step d) is optional insofar as, when the process is used several times under the same conditions, the experimenter knows the cell types present at the different stages of culture and their relative proportions without having to repeat the step of identification.
  • the method further comprises the implementation of depletion techniques or enrichment before the culturing step c) or before expansion, in order to modify the proportions of the different cell types.
  • cell population or “cell population” mean any population of non-pure cells, generally containing a dominant cell type and one or more minority cell types.
  • the dominant cell type is the cell type whose proportion in the cell population is the highest.
  • a dominant cell type is the cell type whose proportion in the cell population exceeds 50%.
  • a cell therapy product suitable for human administration comprises an isotonic solution in which the cells are resuspended. This solution must be free of toxic components present in the freezing media. Such a component is, for example, DMSO.
  • the invention results in particular from the observation that the method makes it possible to obtain a population of cells whose composition is adapted to the desired therapeutic effect. It makes it possible to obtain a population of cells whose dominant cell type expresses the CD56 + marker and the HLA class I marker without prior cloning, nor positive selection of the cells expressing the CD56 + marker.
  • the muscular biopsy is usually performed by collecting cubes of 2 to 4 cm side. From 0.05 grams to several tens of grams can be taken as needed.
  • One of the advantages of the method is that it makes it possible to obtain a very large number of cells of a cell type in a dominant proportion, varying from a few thousand to several billion, depending on the cell type sought, the number of expansions carried out, and the time allotted for each passage.
  • the method makes it possible to produce up to several hundred million cells expressing the CD56 antigen within two to three weeks. The process allows for many phases of expansion, allows the theoretical achievement of at least 100 billion cells expressing CD56 + antigen and HLA class I antigen after 8 to 9 expansions.
  • the muscular tissue taken is a skeletal muscle tissue, preferentially taken from the adult, the young adult, the adolescent or the child.
  • the muscle tissue removed is fetal skeletal muscle tissue.
  • the cells can be obtained especially from the vastus externa, vastus interna, biceps, quadriceps, hamstrings, gastrocnemius, peroneal, deltoid, dorsal, sternocleidomastoid, intercostal, homo-hyoid, right or psoas.
  • the slicing consists in cutting the biopsy into sections of a size preferably less than 0.5 mm placed in a suitable culture medium. Slicing is an essential step to enable effective subsequent enzymatic dissociation. Emining can be done manually using fine scissors. Unexpectedly, however, it has been found that when the slicing step is carried out in an assisted manner, for example using knife crushers powered by electrical or mechanical energy, the method of the invention in which the culture medium is adapted to myoblast differentiation makes it possible to obtain a population whose percentage of cells expressing the CD56 antigen is particularly high.
  • An example of such a usable crusher is the Medimachine ® mill (distributed by Becton-Dickinson).
  • the method of the invention is characterized in that the slicing step is assisted by mechanical or electric knife mills.
  • Muscle tissues consist of muscle fibers.
  • the satellite cells are located under the basal lamina of the muscle fibers.
  • the step of dissociation of the muscle fibers and detachment of the satellite cells is therefore a necessary step for their isolation.
  • the dissociation step consists in the use of digestion enzymes of the extracellular matrix, it can be completed by a mechanical dissociation by suction and delivery of the suspension through a pipette.
  • the dissociation step is in two stages; a first incubation in the presence of collagenase and a second incubation in the presence of trypsin.
  • a first incubation in the presence of collagenase particularly effective levels of use in the emine are between 0.05 to 2 mg / ml.
  • the incubation time at 37 ° C. for such concentrations is then chosen in a time range extending from 15 minutes to 2 hours.
  • the activity of dissociation enzymes is neutralized after dissociation or detachment of the cell layer in order to avoid cell damage.
  • a filtration step will be carried out in two steps: a filtration step on a 100 ⁇ m sieve, then a second step on a 40 ⁇ m sieve.
  • the cells collected after filtration are transferred to a culture reactor in the presence of a medium whose composition allows their growth and / or their differentiation.
  • the composition of the medium is chosen according to the dominant cell type desired at the end of culture.
  • some of the preparation can be frozen. This can be a part of the initial preparation or a subpopulation from an enrichment or depletion stage.
  • the culture media used contain one or more growth and / or differentiation factors whose role is to direct the progenitor cells to a specific differentiation pathway and to proliferate them.
  • growth factors By way of example of growth factors, mention may be made of fibroblast growth factors, bFGF, aFGF, FGF6, hepatocyte growth factor, HGF / SF, epidermal factor, EGF and the various factors characterized such as IGFs. -1, PDGF, LIF, VEGF, SCF, TGFb, TNFa, IL-6, IL-15, NGF, neuregulin, thrombopoietin and growth hormone.
  • IGFs -1, PDGF, LIF, VEGF, SCF, TGFb, TNFa, IL-6, IL-15, NGF, neuregulin, thrombopoietin and growth hormone.
  • hormones or active molecules that can be used in the composition of media, such as glucocorticoids (natural or semi-synthetic, ie hydrocortisone, dexamethasone, prednisolone or triamcinolone), progestagens and derivatives ( progesterone), estrogens and derivatives (estradiol), androgens and derivatives (testosterone), mineralocarticoids and derivatives (aldosterone), LH hormones, LH-RH, FSH and TSH, thyroid hormones T3, T4, retinoic acid and its derivatives, calcitonin, prostaglandins E2 and F2 / alpha or parathyroid hormone.
  • glucocorticoids natural or semi-synthetic, ie hydrocortisone, dexamethasone, prednisolone or triamcinolone
  • progestagens and derivatives progesterone
  • estrogens and derivatives estradiol
  • androgens and derivatives testo
  • the preparation Prior to culturing, or during an expansion phase, the preparation may be enriched or depleted. These operations are carried out by those skilled in the art using the various techniques proposed in the state of the art to operate a selective sorting. These techniques rely on the identification of extracellular antigens characteristic of this or that cell type by specific reagents.
  • CD34 and CD56 antigens expressed on certain cells present within the muscle cell population may be used. Initial sorting of the total cell population according to the expression of these two antigens thus makes it possible to separate two groups. In particular, it makes it possible to separate CD34 + cell types from CD34 cell types. It has been shown that the CD34 + population generates a dominant CD15 + cell type, CD56- not expressing desmin.
  • the CD34 population generates a dominant CD56 +, CD15-expressing desmin cell type.
  • the population CD34- generates a population CD56-, CD15-.
  • the method of the invention comprises a stage of depletion of CD34- or CD34 + cells leading respectively to a population comprising a dominant cell type C015 + or CD56 +.
  • One embodiment of the method of the invention therefore relates to a method for obtaining a population of cells of which a dominant cell type is the myoblastic cell type, characterized in that it comprises a stage of depletion of CD34 + cells before the cell culture or during one of the expansion phases.
  • the culture reactor is preferably static. It must have a large culture surface compared to conventional carriers (petri dishes, flasks) so as to harvest in a few days a large cell population.
  • An example of such a culture reactor is the tray culture device (single, double and / or multi-stage).
  • the culture device used in the method also allows sterile sampling of the cells in order to carry out the samples necessary for the identification of the cell types present at the different stages of the culture by analysis of specific markers. It allows the emptying of media, the washing and detachment of cells and finally their harvest in a sterile manner.
  • bags are used and specially adapted sterile tubings connect the bags to the reactor to allow transfer of media or cell harvesting.
  • This device thus makes it possible to perform a large number of operations in a closed system.
  • the number of days of culture varies from 0 to 45 days.
  • the culture can be continued by conventional expansion or perfusion techniques for a period of up to several months.
  • the expansion phases comprise a step of detaching the cells, washing the cells and restoring culture to a larger culture surface, the solutions and enzymes used to carry out these steps being well known to those skilled in the art.
  • the method of the invention comprises at least three phases of cell expansions. Such a method makes it possible to multiply the number of cells without substantially modifying the proportions of the cell types obtained at the end of culture of each expansion.
  • Kinetics of differentiation is carried out in the method of the invention by identifying cell types present in the cell populations obtained at different stages of the culture.
  • characterization of cell populations This characterization is carried out from samples taken during the cultivation, during the culture and during the harvesting of the cells.
  • the characterization of the cell populations can also relate to a cell culture carried out in parallel on a smaller scale but under identical or equivalent conditions in terms of culture medium and method of carrying out the expansion phases.
  • the characterization relates to the adherent cells or the cells present in the supernatant.
  • the different stages of the culture are counted in days starting from the day of culture in a reactor until the appearance of the first differentiated cells or obtaining a degree of confluence of the cells of at least 20 to 50%.
  • the function of cell population characterization is to identify all cell types and their proportions according to the stage of culture. In particular, it makes it possible to highlight the dominant cell types according to the stage of the culture.
  • the method of the invention thus provides a means of selecting a cell population according to the characteristics of the different cell types present in the population.
  • cellular markers denotes any cell antigen that can provide information alone or in combination with other markers on a cell type. Any type of cell marker can be used by the method, the choice of markers used will be guided by the choice of cell types that one wishes to characterize.
  • Any technique capable of characterizing a cellular antigen can be implemented in the method of the invention.
  • Western Blot or immunocytochemistry may be mentioned without being limiting.
  • the techniques allowing the characterization of messenger RNAs encoding said antigens can be used in an equivalent manner.
  • the cellular markers analyzed for the identification of cell types are specifically chosen from the following markers: CD10, CD13, CD15, CD16, CD34, CD38, CD40, CD44, CD45, CD56, CD71, CD80, CD117 or the following structures, CD138, HLA-Class I, HLA-class II, VLA3, VLA5, VLA6, ICAM-1, VCAM and desmin.
  • the markers CD10, CD13, CD15, CD34, CD44, CD56, CD117 and HLA Class I and desmin are used. These markers are identified by the use of specific monoclonal antibodies.
  • Table 1, hereinafter provided in the experimental part indicates, for each of the markers, the cell types which conventionally express the corresponding antigens. It should be noted that these cell markers were initially developed for the characterization of the immuno-hematological system. An originality of the invention consists in the use of these markers for the characterization of cells of muscular origin at the different stages of culture or expansion.
  • the method of the invention is therefore implemented through the analysis of cellular markers not conventionally used in the characterization of cells derived from muscle tissue.
  • cell markers are, for example, CD10, CD13, CD15, CD34, CD44, CD45, CD117 and HLA Class I.
  • the implementation of the method of the invention allows the demonstration of an evolution over time of the dominant cell type present in the culture.
  • the majority populations are those expressing markers of marrow progenitor cells and cells of the lymphoblastoid system. More precisely, a majority of CD34 + / 45- phenotype cells were observed on day 0 and day 1, as well as the presence of a CD117 + minority cell type in the non-adherent cell population.
  • the evolution is then marked by an increasing increase in the proportion of CD15 + and / or CD56 + cells, and a decrease in CD34 + populations.
  • the dominant cell type remains CD34 + from D0 to D5. A gradual transition from a dominant cell type CD34 + to CD56 + cell is observed. On the other hand, the proportion of CD13 +, CD44 +, CD71 + cell types increases with time. Minority populations CD138 +, HLA-Class II + and CD38 + are also observed. At the end of the culture, the dominant cell type is of CD56 + phenotype, especially from the J5 stage until the end of the culture. CD56 + cells also express CD10, CD13, CD44, desmin, HLA-class I, CD44, VLA-3, VLA-5 and VLA-6. These markers characterize myoblastic cells. A second predominant cell type consists of cells expressing CD15, CD10, CD13 and HLA-Class I.
  • a population CD56- / CD15- / CD13 + is also present in variable proportions constituting a third predominant cell type.
  • this population one part expresses the desmin and one part does not express it.
  • the implementation of the method of the invention has thus made it possible to distinguish three cell types present in the cell populations, the respective proportions of which change significantly during the course of the culture, the CD34 + cells, the CD15 + cells and the CD56 + cells. .
  • the characterization of the cell populations consists in determining the respective percentage of the three CD34 +, CD15 + and CD56 + cell types at the different stages of the culture.
  • a desired cell type in a cell population is separated after selection of the culture stage during which the desired cell type is in the desired qualitative or quantitative state, particularly after selection of the culture during which the desired cell type is dominant.
  • the presence in predominant proportion of the cells to be separated facilitates their preparation.
  • the invention thus provides a method for obtaining a cell population of high purity.
  • high purity cell population is meant a population of cells in which the dominant cell type is in excess of 50% in the cell population. It is clear that one skilled in the art can implement the various techniques proposed in the state of the art for a selective sorting of said cells. By way of example, mention may be made of cloning, flow cytofluorometry or immunoaffinity or immunomagnetic columns using antibodies specific for the cells in question.
  • the method of the invention is characterized in that it does not include purification steps, positive selection, or cloning of a specific cell type.
  • positive selection it is necessary to understand any step for sorting the cells on the basis of at least one particular characteristic and in particular the expression of a cellular marker.
  • the method of the invention unlike the methods described in the state of the art, makes it possible to obtain a cell therapy product comprising a dominant myoblast-type cell type, without a preferential selection step. of the myoblastic cell type.
  • the absence of cloning steps, purification or positive selection significantly improves the final yield obtained after the expansion procedures in terms of numbers of cells obtained.
  • an aliquot of cells can be removed and cultured separately.
  • the medium may be the same or of different composition.
  • the culture media are selected according to the desired differentiation pathway.
  • An example of a medium for differentiation into myoblasts, endothelial cells, smooth muscle cells or myofibroblasts is the MCDB 120 medium supplemented with fetal calf serum (23) which in particular comprises a glucocorticoid such as dexamethasone and bFGF.
  • Another example of a preferred medium is the modified MCDB 120 medium substituting L-valine for D-valine. It has indeed been found that such a modification makes it possible to obtain a particularly selective medium for the differentiation of the cells into myoblasts.
  • Such a medium makes it possible to obtain, with the method of the invention, a population comprising a very large number of cells, most of which are of the CD56 + or HLA Class 1 type.
  • a differentiation medium in adipose tissue contains, in particular, dexamethasone, isobutylmethylxanthine and, if appropriate, serum fetal calf, indomethacin and insulin. Maintaining differentiation is achieved in medium containing 10% fetal calf serum and insulin.
  • the cells are centrifuged in micromass and cultured in medium without serum but containing TGF-beta3.
  • a bone tissue differentiation medium contains dexamethasone, beta-glycerol phosphate, ascorbate, and, where appropriate, fetal calf serum.
  • the cells are harvested at the selected culture stage depending on the cell population that is desired.
  • Non-adherent cells present in the supernatant and / or adherent cells can be harvested by this method.
  • the cell populations present in the supernatant and adherent cells may be of different composition. Consequently, the mode of harvesting the cells will be a function of the desired cell population.
  • the harvesting of the adherent cells is carried out by enzymatic dissociation of the cells and detachment of the cell layer by techniques known to those skilled in the art.
  • the non-adherent cells are harvested by aspiration.
  • the implementation of the above method including the establishment of kinetics of differentiation from cells derived from muscle tissue biopsies allows obtaining characterized cell populations.
  • the cell populations can be obtained by a method similar to the method for obtaining cell populations described above, but not including a step of identifying the cell types at different levels. cellular stages. Indeed, the step of identifying the cell types is necessary for the choice of the harvesting stage. Since, having carried out at least once the process of the invention, the skilled person knows the optimal stage of harvesting cells according to the desired population, it can obtain the same types of populations by limiting the number of markers used for the characterization of cell populations and the stages at which it performs this characterization. Thus, by "populations of cells that can be obtained", it is therefore necessary to include a population of cells obtained by the process of the invention not necessarily comprising a step of characterizing cell populations within the meaning of the invention, such as as described above.
  • the optimal harvest stage is known by implementing a differentiation kinetics on a prior culture, carried out under the same conditions.
  • the different cell populations that can be obtained by the method of the invention, with or without an identification step, are of different types depending on the culture stage chosen.
  • the invention provides a cell population whose dominant cell type expresses the CD34 marker.
  • the cell population also includes a minority cell type of CD117 + phenotype.
  • the invention provides a cell population whose dominant cell type expresses the CD15 marker.
  • a preferred embodiment of the invention provides a cell population whose dominant cell type is myoblastic cells.
  • Myoblastic cells are characterized by the analysis of CD56 marker expression. They are preferably characterized by CD56 marker expression analysis in combination with the analysis of CD10, CD13, CD44, HLA Class I and desmin markers. Analysis of the expression of desmin, an intracellular cytoskeletal protein specific to myoblastic cells and differentiated muscle cells requires a suitable FACS labeling and analysis protocol, detailed in the experimental section.
  • the cell population additionally comprises a doubly negative population CD56 - / CD15 - .
  • the invention provides in particular a cell population derived from the same muscle biopsy and comprising from 50 ⁇ 10 6 to 800 ⁇ 10 9 cells, preferably at least 500 ⁇ 10 6 cells, of which at least 50% of the cells, and better still at least 60%, and more preferably at least 70%, are CD56 +.
  • the invention relates to the use of a population of CD56 + cells obtained according to the methods described above in the preparation of a cellular therapy product for the treatment in humans of post-ischemic heart failure or for repair of heart tissue.
  • the implementation of the method makes it possible to obtain a large number of cells rapidly and the cell populations obtained have the advantage of being homogeneous, and therefore particularly suitable for the preparation of a cell therapy product.
  • It also relates to the use of a population of cells from the same muscle biopsy and comprising from 500x10 6 to 800 x 10 9 cells of which at least 50%, and better still at least 60%, are CD56 + or HLA Class 1 in the preparation of a cellular therapy product for treatment in humans, post-ischemic heart failure or any genetic, viral, medicinal, infectious or parasitic heart disease
  • the treatment of heart disease consists in particular of injecting with a needle a population of cells, the dominant type has the characteristics of myoblastic cells, obtained and prepared as a cell therapy product, directly in the myocardial tissue (12) or indirectly in the arterial circulation (24).
  • At least 600 x 10 6 cells from the same biopsy are injected.
  • Heart failure is now managed by treatment with angiotensin converting enzyme (ACEI) inhibitors.
  • ACEI angiotensin converting enzyme
  • the invention therefore also relates to the use of cell populations of muscle origin obtained according to the methods described above as a transplant for potentiating the pharmacological treatments of heart failure.
  • the experimental part of the present text describes the carrying out of autologous muscle cell transplantations in rats demonstrating the feasibility of this technique. Indeed, the results show that the transplantation of these cells in the rat significantly improves the functional evaluation parameters thus demonstrating the feasibility of such transplants. They also show that the improvement in myocardial function is related to the number of grafted cells.
  • the experimental part also describes the results obtained in the rat, during the joint use of autologous muscle cells and the pharmacological treatment of heart failure.
  • a step of genetic modification of the cells by transfection of a heterologous nucleic acid can be performed.
  • the nucleic acid is selected to allow expression of a polypeptide or protein in the transfected cells.
  • the transfected cells are then transplanted and allow delivery of the polypeptide or protein expressed from the heterologous nucleic acid, wherein said polypeptide or protein is a biologically active product.
  • the invention thus relates to the use of a cell population as a cell therapy product as a delivery platform for a biologically active product.
  • the first part presents the examples of implementation of the method according to the selected culture stage for obtaining cell populations whose dominant cell type is CD34 + or CD15 + or CD56 + (myoblastic) or double negative CD56- / CD15-.
  • results presented in the second part show the efficiency of the technique of transplantation of myoblastic cells on tissues cardiac infarction in the rat. It also makes it possible to determine the criteria required for a good efficiency.
  • a third part presents human clinical trials of muscle cell transplantation for myocardial tissue reconstitution, myocardial tissue repair, metabolically active tissue generation, tissue generation with functional activity. does not exist before reconstitution. It also shows that muscle cells can also play a role in remodeling heart tissue in humans.
  • Medium A Modified MCDB120 medium (23): substitution of L-Valine with D-Valine, removal of phenol red, removal of thymidine.
  • Medium B Medium A + 20% irradiated fetal calf serum + antibiotics (at 100 IU / ml for penicillin and 100 ⁇ g / ml for streptomycin).
  • Medium C Medium B + bFGF (10 ng / ml) + dexamethasone (1 ⁇ M).
  • Medium E Medium A + bFGF (10 ng / ml) + dexamethasone (1 ⁇ M) + sterile human serum albumin (0.5%).
  • Solution F Sterile isotonic saline solution 0.9% NaCl.
  • Solution G Solution F + 4% human serum albumin and 7.5% final DMSO (dimethyl sulfoxide).
  • Solution H Injection solution F + 0.5% human serum albumin
  • the cells are centrifuged at 160 g for 5 minutes. Cell counting and population analysis are performed using a Neubauer hemacytometer. In the expansion phases, the cells are incubated at 37 ° C. in an air-CO 2 incubator (95% -5%) saturated with moisture. The cells are observed using an inverted phase-contrast microscope.
  • the sample is taken in the operating room, in a sterile environment, in an open system.
  • a biopsy of approximately 10-16 grams of skeletal muscle tissue is performed by the surgical team.
  • the biopsy is then cut into small cubes of 2 to 4 mm on one side, then minced with fine scissors in medium A.
  • the emine is placed in a sterile bottle containing 25 ml of medium A.
  • the slicing stage is also assisted, using Medimachine ® knife mills (distributed by Becton-Dickinson).
  • Medimachine ® knife mills distributed by Becton-Dickinson.
  • fragments of mass less than 0.2 g are dissociated in the sterile Medicon receptacles, after grinding controlled by an electric motor, lasting less than 5 minutes.
  • Repetition of the operation using several receptacles Medicon allows to finally prepare several grams of muscle.
  • Table A below shows the proportions of the CD56 +, CD15 + and CD34 + cell types obtained during the different culture stages J0, J15, J20 and J26 (the proportion of CD34 cells being almost zero at J15, this one does not is not indicated in the table for the steps following the cultivation J0).
  • the biopsy cutting step is a crucial step in rapidly obtaining a population comprising a dominant CD56 + myoblast type cell type.
  • Biopsies were taken from patients aged 15 to 73 years. The results presented in Table D below show that the method is applicable regardless of the age of the patient on which the biopsy is taken.
  • Table D Preparation of muscle-derived cells from biopsies from patients of different ages Day 0 First passage Second passage Third passage Last name Weight (g) Patient age (years) Number 10 * 5 % CD56 + Number 10 * 6 % CD56 + Number 10 * 6 % CD56 + Number 10 * 6 % CD56 + MYO1 14.9 73 100 3.2 19.5 48 315 58.3 890 67.3 MYO3 10.4 63 43 3.4 14.2 67.7 156 87.1 922 91.3 MYO4 13.9 67 102 32.3 3.4 76.9 115 97.5 657 97.1 MYO5 11.6 39 117 22.1 31 71.5 244 89.9 993 95.2 MYO6 12 55 164 28.7 26.5 82 483 91.3 1210 84.9 4929 0.19 15 1 ND 1.6
  • Biopsies can be stored for 90 hours at 4 ° C or frozen in balanced saline before being cultured.
  • Tables E and F show that the viability of the cells and the evolution of the proportions of the different types of population born are not significantly affected after 90 hours of preservation of the biopsy at 4 ° C., or after freezing.
  • Table E Placement of a preserved biopsy 90 h at + 4 ° C in a balanced saline solution and preparation of muscle cells according to the method WEIGHT: 1.05 g D0 PASSAGE 1 (J7) CNT 10 * 6 0.8 4.9 % CD34 + 7.7 NS % CD56 + 6.5 58.7 % CD15 + 8.4 37.7 % VIABILITY 90.5 95 NS: not significant PASSAGE 1 PASSAGE 2 PASSAGE 3 BIOPSY J TREATMENT D0 J18 J20 J25 4929 CNT 10 * 5 0.97 16.2 47.3 55?
  • the culture method can be implemented from biopsies of healthy subjects or patients with pathology.
  • the following results presented in Table G show in particular that the the invention can be implemented for the preparation of cells of muscular origin derived from a patient suffering from Duchenne muscular dystrophy.
  • Table G Culture from a thawed biopsy from a patient with Duchenne muscular dystrophy PASSAGE 1 PASSAGE 2 PASSAGE 3 PASSAGE 4 PASSAGE 5 BIOPSY STADIUM D0 J7 J14 J21 J26 J29 4964 CNT 10 * 5 1.58 1.78 11.6 473.6 2411.2 6397 DE 24 H % CD34 + 53.9 ND 0.3 0 ND 0 striated muscle % CD56 + 5 24.6 78.1 73.7 60 52.5 paravertébral % CD15 + 0.4 30.4 12.7 15.5 26.4 36.1 WEIGHT: 0.136 g % VIABILITY ND 94 82.5 91.2 95.6 98
  • biopsies were obtained from different muscles, the paravertebral, the anterior tibialis, the peroneal longus, the common extensor toes, the peroneal longus, the posterior tibialis and the soleus.
  • the results obtained according to different biopsies are presented in Table H.
  • the vial containing the emine is centrifuged at room temperature. The supernatant is removed by aspiration. The weight of the emerald is obtained by weighing on a weighed scale using an empty bottle. The emulsion is rinsed with 25 ml of medium A. After sedimentation of the emulsions, the supernatant is removed by suction.
  • a liberase solution (Roche-Boehringer) was prepared according to the manufacturer's instructions and then reconditioned and frozen at a concentration of 10 mg / ml.
  • the liberase is defrosted extemporaneously and then added to the emulsion at a concentration of 0.1 mg / ml, in a volume of 10 ml per gram of tissue.
  • the bottle is placed in an oven at 37 ° C for a period of 60 minutes.
  • the bottle is agitated manually every 5 to 10 minutes (diffusion of the enzyme and gentle mechanical dissociation).
  • the suspension is then centrifuged.
  • the supernatant is removed by pipetting. This first digestion product is then incubated in a 0.25% trypsin solution.
  • the digestion product is filtered through a 100 ⁇ m sieve and then 40 ⁇ m under gravity pressure in order to separate the dissociated cells from the residual tissues.
  • One sieve per gram of tissue is used (Falcon Cell strainer).
  • the filtrate is centrifuged at 300 g for 5 minutes. After removal of the supernatant, the pellets are washed with medium B and then centrifuged. The supernatant is removed by aspiration. The pellet is then taken up in 10 ml of medium C. 100 ⁇ l of volume is taken for counting. An aliquot is reserved for estimating viability by cytofluorimetry (propidium iodide).
  • the culture device is a culture tray (Nunc Single Tray) with an area of 600 cm 2 .
  • the filling of the tray is done through a hole provided for this purpose and closed by a sterile single-use stopper.
  • the cultures are incubated at 37 ° C. in a chamber saturated with humidity in an air-CO 2 controlled atmosphere (95% -5%).
  • a first emptying is carried out in order to eliminate the dead cells and the muscular debris.
  • An empty bag is connected to one of the two orifices of the culture device.
  • the medium is removed by gravity and replaced with 120 ml of medium C which are added to the culture.
  • Medium C is renewed after 120 to 192 hours.
  • the expansion is decided when the degree of confluence of the cells reaches 20% to 50% or when the first myotubes appear (approximately 8 days after the culturing).
  • the cells are washed by gentle manual stirring with 50 ml of solution D.
  • the solution D is drained and then 20 ml of irradiated trypsin solution (0.25%) are added.
  • the flask is incubated for 5 minutes at 37 ° C.
  • the cells are harvested in a 40 ml bag.
  • the action of trypsin is neutralized by the addition of 10% fetal calf serum.
  • the serum is injected inside the bag using a syringe.
  • the cells are centrifuged.
  • the supernatant is removed by transfer to another emptying bag, connected by a sterile connector.
  • the pellet is resuspended in 30 ml of medium C for washing the cells and then the cells are centrifuged.
  • the supernatant is removed by transfer to another emptying bag, connected by a sterile connector.
  • the cell pellet is resuspended in 20 ml of medium C.
  • An aliquot is taken for counting and analysis of the populations. Viability is estimated using a cytofluorimeter.
  • the cells are then transferred to a bag containing 500 or 750 ml of medium C, then seeded in two or three double-tray units (Nunc double-tray) with a total surface of 1200 or 1800 cm 2 .
  • the cells are incubated.
  • a third expansion is decided when the degree of confluence of the cells reaches 60% to 70% or when the first myotubes appear.
  • multi-stage boxes of 10 trays are used.
  • the cells are washed with 100 ml of solution D.
  • the detachment of the cell layer and the enzymatic dissociation of the cells are carried out after draining the washing solution by the addition of 50 ml of trypsin irradiated (0.25%) in each tray.
  • the preparations are incubated for 5 minutes at 37 ° C.
  • the cells are harvested in sterile pockets of 300 to 600 ml.
  • the action of trypsin is neutralized by the addition of 10% by volume of fetal calf serum.
  • the cells are centrifuged and the supernatant is removed by connection to a drain bag.
  • the cell pellet is resuspended in 50 ml of medium C and then transferred into one or two vials containing 1200 or 2400 ml of medium C by a sterile connector.
  • One or two culture dishes of 10 multi-stage trays (multi-tray Nunc) is seeded with 1200 to 2400 ml of the cell preparation. Visual control of growth in multi-stage trays is impossible. Therefore, a single sterile Nunc culture dish (single-tray) is also seeded with 110 ml of the cell preparation. This last plate allows a daily visual control of the culture and the state of confluence of the cells. Cells are then incubated. The culture lasts 3 to 5 days. The day before harvesting the cells, the medium is removed by emptying into a sterile bag and replaced by an equivalent volume of medium E. The final harvest is decided when the degree of confluence of the cells reaches 90% or when the first myotubes appear.
  • Characterization is based on flow cytofluorometric analysis (FACS).
  • Antibodies to the following human cell surface antigens were used: CD5, CD10, CD11, CD13, CD14, CD15, CD16, CD18, CD19, CD20, CD28, CD31, CD34, CD38, CD40, CD40-ligand, CD44 , CD45, CD56, CD62, CD71, CD80, CD86, CD90, CD105, CD117, CD138.
  • Antibodies directed against the following antigenic structures have also been used: CD138, HLA-Class I, HLA-DR, ELAM, ICAM, LECAM, Stro-1, S-endo-1, VCAM, VLA2, VLA3, VLA4, VLA5, VLA6.
  • Table I Presentation of certain cellular markers used in the process and the cell types expressing them.
  • MARKER PEN CELLULAR TYPES CD10 preB lymphocytes. neutrophil CD13 monocytes, myeloid cells CD15 monocytes, macrophages, granulocytes, eosinophils CD16 NK, under pop. T lymphocytes, neutrophils CD34 progenitors CD38 LT activities, stem cell, under pop. L T, B, NK CD40 activated CD4 + T cells CD44 Anti HCAM CD45 leukocytes CD56 NK, under pop.
  • Table JA Identification of cell types over time (as a percentage in the total cell population) of stages J0 to J3 of the culture.
  • the method makes it possible to obtain at 3 ⁇ 10 5 to 4 ⁇ 10 6 cells per gram of tissue.
  • the majority cell types are CD34 +.
  • the CD34 + cell types are CD34 + / CD10 + at 14%; CD34 + / CD10- at 25%; CD34 + / CD56 + at 0%; CD34 + / DR + at 14% and CD34 + / DR at 28%.
  • the CD34 + populations are predominantly CD45-. Minority populations express CD44, CD45, CD56, CD117, HLA-Class II.
  • CD34 + A majority of cells is CD34 +.
  • the population consists of CD34 + / CD10 + (27%) and CD34 + / CD10- (47%). CD13 appears.
  • the preparation is negative for CD117 and CD45.
  • the CD15 + and CD56 + cell types are in the minority.
  • CD34 + A significant proportion of cells is CD34 +.
  • the population consists mainly of CD34 + / CD10-.
  • a CD117 + population ( ⁇ 5%) is present and expresses CD45 +.
  • Some minority populations are present: CD38 + (15%), CD45 +, few CD15 + and CD56 +, HLA-Class II +.
  • CD15 + and / or CD56 + cells The evolution is marked by an increasing increase in the proportion of CD15 + and / or CD56 + cells and a decrease in CD34 + populations. The gradual transition from a CD34 + to CD15 + population can be observed over time. The proportion of CD13 +, CD44 +, CD71 + populations increases with time. There are minority populations CD138 +, HLA-Class II +, CD38 +.
  • CD56 +, CD15 + and CD56- / CD15- there are three predominant populations: CD56 +, CD15 + and CD56- / CD15-.
  • the CD56 + population expresses CD10, CD13, CD44, desmin and HLA-Class I. These markers are specific for myoblastic cells.
  • the CD15 + population expresses CD13, Class I and partially CD10.
  • the CD56- / CD15- population one fraction expresses desmin and the other fraction does not express it.
  • Some markers are expressed more weakly and variably: CD71, VLA3, VLA5, VLA6, CD16 + and CD40L.
  • the CD34 +, CD38 +, CD45 + and HLA-Class II + populations have disappeared or are extremely minor.
  • Table K represents the characteristics of the cells obtained at the end of the first pass, by comparing different initial conditions. Eight independent experiments are represented. After depletion of the CD34 + fraction, the method makes it possible to rapidly obtain a cell population that is predominantly composed of cells expressing CD56. In particular, the proportion of cells expressing CD56 is greater than that obtained from an unexplained biopsy. ⁇ u> Table K. Depletions or enrichments in CD34 + cells present in muscle biopsy.
  • the following protocol describes harvesting the cells at the final stage to obtain a majority myoblastic population.
  • the protocol allows the person skilled in the art to implement it whatever the stage of differentiation chosen for harvesting the cells and this, depending on the desired cell population.
  • the cells are washed with 500 ml of solution D (for the multi-tray units), 50 ml for the single unit and 100 ml for the double units.
  • the wash solution is drained and 200 ml of irradiated trypsin solution (0.25%) is added to the multi-tray units (20 ml for the single unit, 40 ml for the double units).
  • the preparation is incubated for 5 minutes at 37 ° C.
  • the cells are collected by emptying in a 500 ml bag.
  • the action of trypsin is neutralized by the addition of 10% fetal calf serum injected by a syringe.
  • the cells are washed as follows: the cells are centrifuged.
  • the supernatant is removed and the cells are resuspended in 300 ml of solution F and centrifuged. Supernatants are eliminated. Two other washing steps as previously described are carried out. These successive washes are intended to remove trypsin, animal proteins still present and recombinant bFGF. During the third wash, an aliquot is reserved for cell count, viability and cell quality estimation, microbiological quality controls.
  • the cells can be concentrated in solution H so as to obtain a suspension that is adequate for the clinical use requested.
  • the cells After centrifugation, the cells are taken up in a volume of isotonic solution at a concentration of 1.5 ⁇ 10 8 cells / ml. They are finally sucked into a 10 ml syringe. The cells are removed for injection into sterile syringes.
  • the type of needle used for the injection depends on the targeted tissue. For direct intramyocardial injection, a 90 ° 25 to 30 gauge bent needle is specifically used.
  • Table L summarizes the results obtained during the implementation of the method of the invention from different biopsies obtained from three different patients.
  • the cells were initially produced in single, double and multiple plate units until the third included expansion.
  • the expansions were subsequently carried out by duplicating the populations and transplanting them into culture dishes of 25 cm 2 . At each pass, the majority of the cells were used for characterization and counting, while a known number of cells were used for duplicate seeding. The number of cumulative doublings allows, by calculation, to obtain about 100 billion cells between the eighth and the ninth expansion.
  • Table L shows the yields in terms of number of cells obtained and in terms of proportion of CD56 + or desmin + cells in the population at the different expansion phases for the three patients named MYO 003, MYO 004 and MYO 005.
  • the histogram at the figure 7 presents the median expression values of CD56 and CD15 on 8 samples during the different phases of expansions.
  • the cells are suspended in the freezing medium (solution G) and transferred to two sterile freezing bags at a concentration of between 10 7 and 2 ⁇ 10 7 cells / ml or in cryocongesting tubes at a concentration of between 1 x 10 6 and 5 x 10 6 / ml.
  • the freezing is carried out using a device (Digicool or Nicool) ensuring a progressive descent in temperature controlled.
  • the cells are stored in liquid nitrogen until defrosting.
  • Thawing of the cells is carried out in a water bath at 37 ° C. Cell preparations are washed twice with isotonic saline. The rinses are performed by sterile connection to the pockets of isotonic solution and the emptying pockets. An aliquot is reserved for estimating viability and cell quality.
  • Wistar male rats weighing 280 g were anesthetized with ketamine (50 mg / kg) and xylasin (10 mg / kg) and ventilated by the trachea. A thoracotomy was performed. Myocardial infarction is obtained by left coronary ligation using 7/0 polypropylene wire.
  • the length measurements (L) of the long axis of the left ventricle and the plots of the endocardial zones (a) are carried out.
  • the anterior muscles of the right and left tibialis are dissected to separate the tendon and the aponeurotic tissue from the muscle tissue. They are then minced, weighed and dissociated with enzymes using collagenase IA (2 mg / ml, Sigma Chemical Co., St. Louis, MO, USA) for one hour and trypsin-EDTA (0.25%). , GIBCO BRL, Gaithersbueg, MD, USA) for 20 minutes.
  • the cells are harvested by sedimentation (7 min at 1200 rpm) and the enzymatic reaction is neutralized by the addition of 10% fetal calf serum. After passing through a 100 ⁇ m sieve and centrifugation, the supernatant is removed and the cells are resuspended in a medium composed of F12 (HAM) with 20% fetal calf serum, 1% (vol / vol) penicillin-streptomycin (10,000 IU / ml-10000 ⁇ g / ml, GIBCO BRL) and 5 ⁇ g / ml bFGF (Sigma).
  • F12 HAM
  • penicillin-streptomycin 10,000 IU / ml-10000 ⁇ g / ml, GIBCO BRL
  • 5 ⁇ g / ml bFGF Sigma
  • the initial seeding is carried out in culture flasks of 75 cm 2 and the cells are incubated in air saturated with moisture at 5% CO 2 .
  • the cells were harvested by trypsinization, washed and viability tested. A sample was seeded on 12-well dishes in 2.0 ml of culture medium for counting. The cells are washed in the injection medium (culture medium + 0.5% BSA, fraction V) and kept in the ice before transplantation. The cells are centrifuged, resuspended in 150 .mu.l of injection medium and administered sub-epicardially in the infarcted zone.
  • rats Forty-four rats were included in this study and were divided into two groups: a control group and a treated group.
  • the cells seeded in a 12-well dish are fixed with methanol and cooled at -20 ° C. for 5 minutes. Non-specific staining is neutralized using a mixture of 5% horse serum (HS) and 5% fetal calf serum in PBS for 20 minutes.
  • the cells are incubated with a mouse antibody directed against desmin (1/200 DAKO, A / S-Denmark) for 1 hour and then with an anti-mouse antibody conjugated to Cy3 marker (1/200, Jackson Immuno Research Laboratories, Inc. .) for an hour in the dark.
  • the cells are observed using an inverted phase contrast and fluorescent illumination microscope. Several snapshots were taken randomly. The proportion of myoblasts is calculated by dividing the number of desmin-positive cells by the total number of cells observed.
  • the rats are sacrificed by an overdose of ketamine and xylasin.
  • the ventricles are isolated and divided into two parts along their longitudinal axis. Both parts are put in isopentane and frozen with nitrogen. Thin 8 ⁇ m sections are prepared using a cryostat and standard histological studies are performed by hematoxylin and eosin staining.
  • the number of injected cells is 3,500,000 ⁇ 500,000 ranging from 700,000 to 6.5x10 6 .
  • the management of heart failure currently consists of the administration of ACE inhibitors. It was therefore interesting to study whether there could be a synergy between the transplantation of cells of muscle origin obtained according to the method of the invention and the protective effect provided by the ACE inhibitors.
  • Myocardial infarction was performed in 39 rats by ligation of the coronary arteries.
  • Perindoprilat treatment at 1 mg / kg daily, an ACE inhibitor, was initiated immediately after infarction and continued without interruption until the animal was sacrificed.
  • Left ventricular function was tested by echocardiography one month after transplantation.
  • the values of ejection fractions increased in both groups and were 32% ⁇ 2% for control and 38% ⁇ 2% for the treated group.
  • Analysis of the volume data showed that the functional improvement provided by muscle-derived cell transplantation is primarily related to an increase in contractility rather than a change in the left ventricle.
  • PET positron emission tomography
  • the cell culture to be injected is then deposited in a sterile stainless steel cup to be sucked into a 1 ml syringe.
  • a coronary bypass is first performed under extra-corporeal circulation according to the usual technique. After analysis of the extension of the infarct and identification of the edges of the necrotic zone, the cell suspension of approximately 650 to 1200 million cells (1.5x10 8 cells / ml) is injected into and at the confines of the zone. infarction, using the 1 ml syringe. Several injections are necessary to deposit all the cells. This operating time is performed under extracorporeal circulation and clamping.
  • the patient is a 72-year-old man hospitalized for heart failure (NYHA Class III) following an extensive inferior infarction of myocardial infarction that is refractory to medical treatment, especially since beta-blockers and angiotensin-converting enzyme inhibitors have had to be interrupted because not tolerated.
  • NYHA Class III The extracardiac balance also notes a moderate renal insufficiency (creatinine: 200 mmol / L) and a bilateral carotid occlusion without functional repercussion to the intracranial Doppler and therefore not an autonomous vascular gesture.
  • the left ventricular ejection fraction is 20% with extensive akinesia of the lower wall and severe anterolateral hypokinesia.
  • the lack of viability in this lower wall is demonstrated by the persistence of akinesia under low dose dobutamine.
  • the anterolateral wall has a biphasic response to dobutamine (low dose, then high dose) indicating viability and ischemia.
  • PET positron emission tomography
  • FDG 2 fluoro 18 deoxyglucose
  • the myoblasts are produced according to the method of the invention described in part A. The number of cells is multiplied using several expansions in multi-stage trays to obtain in two weeks 800x10 6 cells, 65% of cells CD56 +. The proportion of viable cells is greater than 96%.
  • tissue Doppler imaging shows the appearance of a transmyocardial velocity gradient in systole.
  • a new FDG positron emission tomography clearly shows the uptake of the tracer in the lower wall, with a ratio of activity between wall and septum (taken as a control), which increases from 0.5 before the operation to 0 , 7, which reflects a new metabolic activity in the infarcted zone devoid of preoperative viability ( Figure 6 ).
  • This last observation can not have been influenced by the associated myocardial revascularization and, confronted with the echocardiographic data, suggests that the functional improvement in the infarcted zone is really related to the presence of grafted myoblasts.

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicinal Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Biomedical Technology (AREA)
  • Rheumatology (AREA)
  • Zoology (AREA)
  • Cardiology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Neurology (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Cell Biology (AREA)
  • Vascular Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Diabetes (AREA)
  • Hospice & Palliative Care (AREA)
  • Hematology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

A method for obtaining cell populations derived from the muscular tissue and their use for preparing cell therapy products includes culturing cells previously removed by biopsy from skeletal muscular tissues, identifying the different types of cells present at different stages of culture, selecting the culture stage on the basis of the required cell population and collecting the selected culture stage for preparing a cell therapy product. The invention also concerns cell populations derived from muscular tissue obtained by implementing the method whereof the dominant cell type is CD34+, CD15+ or CD56+ or Class 1+HLA, or comprises a doubly negative CD56−/CD15− cell type or may comprise more minority CD10+, Stro-1+ and CD 117+ cell types.

Description

La thérapie cellulaire est une technique à fort potentiel pour le traitement de nombreuses pathologies. Le principe de la thérapie cellulaire repose sur la possibilité de reconstituer un tissu endommagé ou de restaurer une fonction biologique perdue ou altérée au sein d'un tissu, à partir de cellules spécifiques cultivées ex vivo et transplantées sur le tissu malade. Un autre intérêt de la thérapie cellulaire est la possibilité d'utiliser les cellules transplantées comme plate-forme de délivrance d'un produit biologiquement actif, si besoin après modification génétique des cellules avant transplantation. De nombreux essais de thérapie cellulaire sont décris à partir de cultures primaires de différents types cellulaires. On peut citer les transplantations de cellules neuronales réalisées pour le traitement de la maladie de Huntington (1) ou la maladie de Parkinson (2), les transplantations de cellules d'ilôts de Langerhans réalisées pour le traitement du diabète (3), ou encore les transplantations de cellules myoblastiques réalisées pour le traitement de la dystrophie musculaire de Duchenne (4, 5, 6, 7) ou après modification génétique des cellules, pour le traitement du nanisme (8), de l'hémophilie (9) et de la maladie de Parkinson (10).Cell therapy is a technique with high potential for the treatment of many pathologies. The principle of cell therapy is based on the possibility of reconstituting damaged tissue or restoring lost or altered tissue function within a tissue, from specific cells cultured ex vivo and transplanted to the diseased tissue. Another advantage of cell therapy is the possibility of using the transplanted cells as a delivery platform for a biologically active product, if necessary after genetic modification of the cells before transplantation. Many cell therapy trials are described from primary cultures of different cell types. These include neuronal cell transplants performed for the treatment of Huntington's disease (1) or Parkinson's disease (2), Langerhan's islet cell transplants performed for the treatment of diabetes (3), or Myoblastic cell transplants performed for the treatment of Duchenne muscular dystrophy (4, 5, 6, 7) or after genetic modification of the cells, for the treatment of dwarfism (8), hemophilia (9) and Parkinson's disease (10).

La régénération du muscle squelettique est assurée par les cellules satellites qui sont des cellules myogéniques mononuclées situées sous la lame basale des fibres musculaires. A la suite d'une lésion, ces cellules quittent l'état de quiescence pour entrer dans une phase de prolifération active et prennent le nom de myoblastes. Par la suite, les myoblastes fusionnent pour former les myotubes. Des essais de transplantation de myoblastes ont été réalisés chez l'homme pour traiter la dystrophie musculaire de Duchenne et la dystrophie musculaire de Becker (4, 6, 7, 11). Si l'effet fonctionnel des transplantations décrites dans ces études reste limité, aucun effet secondaire n'a été rapporté en terme d'infection ou de carcinogenèse.The regeneration of skeletal muscle is ensured by the satellite cells which are myogenic myonuclear cells situated under the basal lamina of the muscular fibers. Following an injury, these cells leave the quiescent state to enter an active proliferation phase and take the name of myoblasts. Subsequently, the myoblasts fuse to form the myotubes. Myoblast transplantation trials have been conducted in humans to treat Duchenne muscular dystrophy and Becker muscular dystrophy (4, 6, 7, 11). Although the functional effect of transplants described in these studies remains limited, no side effects have been reported in terms of infection or carcinogenesis.

Par ailleurs, l'utilisation de cellules myoblastiques dans le traitement des cardiopathies, et en particulier de l'insuffisance cardiaque post-isçhémique a été envisagée. En effet, contrairement au tissu musculaire, les tissus myocardiques sont dépourvus de cellules souches capables de former des cadiomyocytes et régénérer des tissus. Le traitement le plus radical de l'insuffisance cardiaque post-ischémique reste à l'heure actuelle, la transplantation cardiaque. Cependant, la pénurie de greffons limite cet usage thérapeutique. La transplantation de cellules issues de tissus musculaires dans le muscle cardiaque a donc été envisagée en tant qu'alternative à la transplantation cardiaque. Des études de transplantation de myoblastes dans le tissu infarci du myocarde ont été réalisées chez le rat, le lapin ou le chien (12, 13, 14). Les résultats de ces études ont montré la faisabilité et une certaine efficacité fonctionnelle de telles transplantations. Des essais de greffes de cardiomyocytes foetaux dans un modèle d'insuffisance cardiaque d'origine iatrogène chez la souris ont aussi montré un certain bénéfice fonctionnel. Cependant, l'utilisation de cellules foetales pose, dans la perspective d'application clinique, de nombreux problèmes éthiques, immunologiques et d'approvisionnement cellulaire. L'utilisation d'une population de cellules myoblastiques issues du muscle squelettique constitue donc une alternative particulièrement intéressante pour la préparation de produit de thérapie cellulaire pour le traitement de l'insuffisance cardiaque post-ischémique, voire pour le traitement de cardiopathies d'origines diverses.Moreover, the use of myoblastic cells in the treatment of heart disease, and in particular of post-ischemic heart failure has been considered. Indeed, unlike muscle tissue, myocardial tissues lack stem cells that can form cadiomyocytes and regenerate tissues. The most radical treatment of post-ischemic heart failure remains at present, heart transplantation. However, the scarcity of grafts limits this therapeutic use. Transplantation of muscle tissue cells into cardiac muscle has therefore been considered as an alternative to cardiac transplantation. Myoblast transplantation studies in infarcted myocardial tissue have been performed in rats, rabbits or dogs (12, 13, 14). The results of these studies have shown the feasibility and some functional efficacy of such transplants. Fetal cardiomyocyte transplant assays in a model of iatrogenic heart failure in mice also showed some functional benefit. However, the use of fetal cells poses, for the clinical application, many ethical, immunological and cellular supply problems. The use of a population of myoblastic cells derived from muscle Skeletal is therefore a particularly interesting alternative for the preparation of cell therapy product for the treatment of post-ischemic heart failure, or even for the treatment of heart diseases of various origins.

L'un des types cellulaires les plus importants contenu dans le tissu musculaire est la cellule satellite, précurseur du myoblaste. C'est le type cellulaire qui a été utilisé dans les différentes études cliniques. Cependant, le tissu musculaire contient d'autres types cellulaires. En particulier, certaines cellules d'origine musculaire pourraient aussi être utilisées pour la reconstitution d'un potentiel hématopoïétique (15, 16). Une étude in vitro a aussi démontré la présence dans le muscle humain de progéniteurs susceptibles de se différencier à long terme en tissus cartilagineux ou osseux (17). Des exemples de milieux permettant la différenciation en tissu adipeux, cartilagineux ou osseux sont décrits pour des cellules souches mésenchymateuses (22).One of the most important cell types in muscle tissue is the satellite cell, a precursor of the myoblast. This is the cell type that has been used in various clinical studies. However, muscle tissue contains other cell types. In particular, certain cells of muscular origin could also be used for the reconstitution of a hematopoietic potential (15, 16). An in vitro study has also demonstrated the presence in human muscle of progenitors that can differentiate in the long term into cartilaginous or bone tissue (17). Examples of media allowing differentiation into adipose, cartilage or bone tissue are described for mesenchymal stem cells (22).

En conséquence, compte tenu des propriétés de différenciation des cellules d'origine musculaire, l'utilisation de ces cellules en thérapie cellulaire apparaît intéressante pour le traitement de nombreuses lésions affectant les tissus du système hématologique et immunologique, les tissus osseux, adipeux, cartilagineux, musculaires ou vasculaires.Consequently, taking into account the differentiation properties of cells of muscular origin, the use of these cells in cell therapy appears interesting for the treatment of numerous lesions affecting the tissues of the hematological and immunological system, the bone, adipose, cartilaginous tissues, muscle or vascular.

Une des difficultés majeures de la thérapie cellulaire reste l'obtention d'une population de cellules suffisamment grande, homogène et dont le degré de différenciation est adapté à l'effet désiré.One of the major difficulties of cell therapy remains obtaining a sufficiently large, homogeneous cell population whose degree of differentiation is adapted to the desired effect.

Des procédés de préparation cellulaire myoblastique et leur utilisation en thérapie cellulaire sont décrits dans l'état de la technique (4, 18, 19, 20, 21, 25, 26, 27). La majorité des procédés comprennent :

  • une étape de prélèvement des tissus musculaires par biopsie,
  • une étape d'éminçage,
  • une étape de dissociation des fibres musculaires par action enzymatique,
  • une étape de séparation des cellules individualisées par filtration,
  • une étape de sélection des cellules myoblastiques par clonage ou par tri cellulaire.
Myoblastic cell preparation methods and their use in cell therapy are described in the state of the art (4, 18, 19, 20, 21, 25, 26, 27). The majority of processes include:
  • a step of collecting muscle tissue by biopsy,
  • a slicing step,
  • a step of dissociation of the muscle fibers by enzymatic action,
  • a step of separating the individualized cells by filtration,
  • a step of selecting myoblastic cells by cloning or by cell sorting.

Afin d'obtenir une population suffisamment dense et riche, voire des populations pures en cellules myoblastiques, il a été proposé de sélectionner les cellules myoblastiques sur la base de l'expression de marqueurs spécifiques. Ainsi, la sélection des cellules myoblastiques peut se faire par le clonage des cellules et la caractérisation ultérieure des clones obtenus par cytofluorimétrie puis sélection des cellules musculaires squelettiques exprimant l'antigène CD56 (7). Une méthode directe de tri des cellules myoblastiques exprimant l'antigène CD56 par cytofluorimétrie de flux et ses avantages pour l'obtention d'une culture pure de myoblastes sont aussi décrits dans l'état de la technique (21).In order to obtain a sufficiently dense and rich population, or even pure populations of myoblastic cells, it has been proposed to select myoblastic cells on the basis of the expression of specific markers. Thus, the selection of myoblastic cells can be done by cloning the cells and subsequently characterizing the clones obtained by cytofluorimetry and then selecting the skeletal muscle cells expressing the CD56 antigen (7). A direct method of sorting myoblastic cells expressing the CD56 antigen by flow cytofluorometry and its advantages for obtaining a pure culture of myoblasts are also described in the state of the art (21).

Les cellules conservées sont ensuite cultivées dans un milieu de culture modifié et spécialement adapté pour la culture des myoblastes (23).The conserved cells are then cultured in a modified culture medium and specially adapted for culturing the myoblasts (23).

La présente invention résulte de l'observation que les cellules issues du tissu musculaire squelettique présentent un potentiel de régénération de nombreux tissus en fonction de leur degré de différenciation. L'invention proposée permet donc de fournir des populations cellulaires bien caractérisées d'origine musculaire.The present invention results from the observation that cells derived from skeletal muscle tissue have a potential for regeneration of many tissues depending on their degree of differentiation. The proposed invention therefore makes it possible to provide well-characterized cell populations of muscular origin.

La présente invention fournit un procédé d'obtention d'une population cellulaire comprenant un type cellulaire dominant, à partir d'une biopsie de tissu musculaire, ledit procédé comprenant les étapes suivantes :

  1. a) prélèvement et éminçage d'une biopsie musculaire ,
  2. b) dissociation enzymatique des fibres et des cellules musculaires et séparation des cellules individualisées par filtration,
  3. c) mise en culture des cellules d'origine musculaire ainsi obtenues dans un réacteur de culture de cellules adhérentes en présence d'un milieu comprenant MCDB 120 ET D-valine substituée à la L-valine suivie le cas échéant d'une ou plusieurs phases d'expansion,
  4. d) identification des types cellulaires présents aux différents stades de la culture par l'analyse de marqueurs cellulaires spécifiques,
  5. e) choix du stade de culture pendant lequel le type cellulaire recherché est en proportion dominante dans la population de cellules,
  6. f) récolte d'une population de cellules au stade de culture choisi en e),
  7. g) le cas échéant, congélation des cellules prélevées à l'étape choisie.
The present invention provides a method of obtaining a cell population comprising a dominant cell type, from a muscle tissue biopsy, said method comprising the following steps:
  1. a) sampling and cutting of a muscle biopsy,
  2. b) enzymatic dissociation of the fibers and muscle cells and separation of the individualized cells by filtration,
  3. c) culturing the cells of muscle origin thus obtained in an adherent cell culture reactor in the presence of a medium comprising MCDB 120 and D-valine substituted for L-valine followed, where appropriate, by one or more phases expansion,
  4. d) identification of the cell types present at the different stages of the culture by the analysis of specific cell markers,
  5. e) choice of the culture stage during which the desired cell type is in a dominant proportion in the cell population,
  6. f) harvesting a cell population at the culture stage selected in e),
  7. g) where appropriate, freezing the cells taken at the chosen step.

L'étape d) est facultative dans la mesure où, lorsque le procédé est utilisé plusieurs fois dans les mêmes conditions, l'expérimentateur connaît les types cellulaires présents aux différents stades de culture et leurs proportions relatives sans avoir à répéter l'étape d'identification.Step d) is optional insofar as, when the process is used several times under the same conditions, the experimenter knows the cell types present at the different stages of culture and their relative proportions without having to repeat the step of identification.

Il a en effet été constaté que l'étape d'identification (d) conduisait à des résultats sensiblement identiques lorsque le même procédé était répété.It has indeed been found that the identification step (d) led to substantially identical results when the same process was repeated.

Dans un mode de réalisation particulier de l'invention, le procédé comprend en plus la mise en oeuvre de techniques de déplétion ou d'enrichissement avant l'étape de culture c) ou avant expansion, afin de modifier les proportions des différents types cellulaires.In a particular embodiment of the invention, the method further comprises the implementation of depletion techniques or enrichment before the culturing step c) or before expansion, in order to modify the proportions of the different cell types.

Les termes « population cellulaire » ou « population de cellules » signifient toute population de cellules non pures, contenant en général un type cellulaire dominant et un ou plusieurs types cellulaires minoritaires. Le type cellulaire dominant est le type cellulaire dont la proportion dans la population de cellules est la plus élevée. Préférentiellement, un type cellulaire dominant est le type cellulaire dont la proportion dans la population de cellules dépasse 50%. Un produit de thérapie cellulaire apte à l'administration humaine comprend une solution isotonique dans laquelle les cellules sont resuspendues. Cette solution doit être exempte des composants toxiques présents dans les milieux de congélation. Un tel composant est, par exemple, le DMSO.The terms "cell population" or "cell population" mean any population of non-pure cells, generally containing a dominant cell type and one or more minority cell types. The dominant cell type is the cell type whose proportion in the cell population is the highest. Preferentially, a dominant cell type is the cell type whose proportion in the cell population exceeds 50%. A cell therapy product suitable for human administration comprises an isotonic solution in which the cells are resuspended. This solution must be free of toxic components present in the freezing media. Such a component is, for example, DMSO.

L'invention résulte en particulier de la constatation que le procédé permet d'obtenir une population de cellules dont la composition est adaptée à l'effet thérapeutique recherché. Il permet d'obtenir une population de cellules dont le type cellulaire dominant exprime le marqueur CD56+ et le marqueur HLA de classe I sans clonage préalable, ni sélection positive des cellules exprimant le marqueur CD56+.The invention results in particular from the observation that the method makes it possible to obtain a population of cells whose composition is adapted to the desired therapeutic effect. It makes it possible to obtain a population of cells whose dominant cell type expresses the CD56 + marker and the HLA class I marker without prior cloning, nor positive selection of the cells expressing the CD56 + marker.

La biopsie musculaire est en général réalisée par prélèvement de cubes de 2 à 4 cm de coté. On peut prélever selon les besoins de 0,05 grammes jusqu'à plusieurs dizaines de grammes. Un des avantages du procédé est qu'il permet d'obtenir un très grand nombre de cellules d'un type cellulaire en proportion dominante, variant de quelques milliers à plusieurs milliards, selon le type cellulaire recherché, le nombre d'expansions réalisées, et le temps alloué pour chaque passage. A titre d'exemple, le procédé permet de produire jusqu'à plusieurs centaines de millions de cellules exprimant l'antigène CD56 dans un délai de deux à trois semaines. Le procédé autorisant de nombreuses phases d'expansion, il permet notamment l'obtention théorique d'au moins 100 milliards de cellules exprimant l'antigène CD56+ et l'antigène HLA de classe I au terme de 8 à 9 expansions. Le tissu musculaire prélevé est un tissu musculaire squelettique, préférentiellement prélevé chez l'adulte, le jeune adulte, l'adolescent ou l'enfant. Dans une forme de réalisation de l'invention, le tissu musculaire prélevé est un tissu musculaire squelettique foetal. Les cellules peuvent être notamment obtenues à partir du vaste externe, vaste interne, biceps, quadriceps, jambiers, gastrocnémiens, péronier, deltoïdes, grand dorsal, sterno-cleido-mastoïdien, intercostal, homo-hyoïdien, grand droit ou du psoas.The muscular biopsy is usually performed by collecting cubes of 2 to 4 cm side. From 0.05 grams to several tens of grams can be taken as needed. One of the advantages of the method is that it makes it possible to obtain a very large number of cells of a cell type in a dominant proportion, varying from a few thousand to several billion, depending on the cell type sought, the number of expansions carried out, and the time allotted for each passage. By way of example, the method makes it possible to produce up to several hundred million cells expressing the CD56 antigen within two to three weeks. The process allows for many phases of expansion, allows the theoretical achievement of at least 100 billion cells expressing CD56 + antigen and HLA class I antigen after 8 to 9 expansions. The muscular tissue taken is a skeletal muscle tissue, preferentially taken from the adult, the young adult, the adolescent or the child. In one embodiment of the invention, the muscle tissue removed is fetal skeletal muscle tissue. The cells can be obtained especially from the vastus externa, vastus interna, biceps, quadriceps, hamstrings, gastrocnemius, peroneal, deltoid, dorsal, sternocleidomastoid, intercostal, homo-hyoid, right or psoas.

L'éminçage consiste à découper la biopsie en sections d'une taille de préférence inférieure à 0,5 mm placées dans un milieu de culture adapté. L'éminçage est une étape essentielle pour permettre une dissociation enzymatique ultérieure efficace. L'éminçage peut être réalisé manuellement à l'aide de ciseaux fins. Cependant, de manière inattendue, il a été constaté que, lorsque l'étape d'éminçage est réalisée de manière assistée, à l'aide par exemple de broyeurs à couteaux mûs par l'énergie électrique ou mécanique, le procédé de l'invention dans lequel le milieu de culture est adapté à la différenciation en myoblaste permet d'obtenir une population dont le pourcentage de cellules exprimant l'antigène CD56 est particulièrement élevé. Un exemple d'un tel broyeur utilisable est le broyeur Medimachine ® (distribué par Becton-Dickinson).The slicing consists in cutting the biopsy into sections of a size preferably less than 0.5 mm placed in a suitable culture medium. Slicing is an essential step to enable effective subsequent enzymatic dissociation. Emining can be done manually using fine scissors. Unexpectedly, however, it has been found that when the slicing step is carried out in an assisted manner, for example using knife crushers powered by electrical or mechanical energy, the method of the invention in which the culture medium is adapted to myoblast differentiation makes it possible to obtain a population whose percentage of cells expressing the CD56 antigen is particularly high. An example of such a usable crusher is the Medimachine ® mill (distributed by Becton-Dickinson).

Par conséquent, dans un mode de réalisation, le procédé de l'invention est caractérisé en ce que l'étape d'éminçage est assistée à l'aide de broyeurs à couteaux, mécaniques ou électriques.Therefore, in one embodiment, the method of the invention is characterized in that the slicing step is assisted by mechanical or electric knife mills.

Les tissus musculaires sont constitués de fibres musculaires. Les cellules satellites sont situées sous la lame basale des fibres musculaires. L'étape de dissociation des fibres musculaires et de décollement des cellules satellites est donc une étape nécessaire pour leur isolement. L'étape de dissociation consiste en l'utilisation d'enzymes de digestion de la matrice extracellulaire, elle peut être complétée par une dissociation mécanique par aspiration et refoulement de la suspension au travers d'une pipette.Muscle tissues consist of muscle fibers. The satellite cells are located under the basal lamina of the muscle fibers. The step of dissociation of the muscle fibers and detachment of the satellite cells is therefore a necessary step for their isolation. The dissociation step consists in the use of digestion enzymes of the extracellular matrix, it can be completed by a mechanical dissociation by suction and delivery of the suspension through a pipette.

Le choix des enzymes et leurs concentrations utilisées pour la dissociation des fibres musculaires et des cellules satellites de l'éminçat est guidé par l'étude de leur efficacité enzymatique, les critères recherchés sont une concentration d'enzyme la moins élevée possible et un temps d'incubation minimal pour une efficacité similaire. Le rendement en cellules obtenus après filtration dépend en partie de la qualité de l'étape de dissociation enzymatique. Des enzymes de digestion utilisables dans le procédé de l'invention seules ou en association sont par exemple :

  • toutes les collagénases, incluant les types IA, S et H partiellement purifiées, ainsi que la forme purifiée commercialisée sous le nom de Libérase par Roche-Boehringer,
  • les trypsines, de toutes origines, en solution dans des tampons contenant ou non de l'EDTA,
  • les dispases (aussi connues sous le nom de protéases),
  • la pronase,
  • les élastases,
  • ou encore, les hyaluronidases.
The choice of the enzymes and their concentrations used for the dissociation of the muscle fibers and the satellite cells of the emine is guided by the study of their enzymatic efficiency, the desired criteria are a concentration of enzyme as low as possible and a time of minimal incubation for similar efficiency. The yield of cells obtained after filtration depends in part on the quality of the enzymatic dissociation step. Digestion enzymes that can be used in the process of the invention, alone or in combination, are, for example:
  • all collagenases, including partially purified IA, S and H types, as well as the purified form sold under the name of Liberase by Roche-Boehringer,
  • trypsins, of all origins, in solution in buffers containing or not containing EDTA,
  • dispases (also known as proteases),
  • the pronase,
  • elastases,
  • or else, hyaluronidases.

De manière préférée, l'étape de dissociation se fait en deux temps ; une première incubation en présence de collagénase et une deuxième incubation en présence de trypsine. Lorsque la libérase est utilisée, des concentrations d'utilisation dans l'éminçat particulièrement efficaces se situent entre 0.05 à 2 mg/ml. Le temps d'incubation à 37°C pour de telles concentrations étant choisi alors dans une gamme de temps s'étalant de 15 minutes à 2 heures. De préférence, l'activité des enzymes de dissociation est neutralisée après dissociation ou décollement de la couche cellulaire dans le but d'éviter l'endommagement des cellules.Preferably, the dissociation step is in two stages; a first incubation in the presence of collagenase and a second incubation in the presence of trypsin. When liberase is used, particularly effective levels of use in the emine are between 0.05 to 2 mg / ml. The incubation time at 37 ° C. for such concentrations is then chosen in a time range extending from 15 minutes to 2 hours. Preferably, the activity of dissociation enzymes is neutralized after dissociation or detachment of the cell layer in order to avoid cell damage.

Après neutralisation de l'activité enzymatique, par addition, par exemple, de sérum de veau foetal, de sérum humain autologue, ou de sérum humain allologue provenant d'un groupe compatible, ou encore d'un inhibiteur d'activité enzymatique, le produit de digestion est ensuite filtré sur tamis, sous pression de la gravité afin d'éliminer les fibres non dissociées et recueillir les cellules décollées des fibres musculaires. De manière préférée, une étape de filtration sera réalisée en deux temps : Une étape de filtration sur un tamis de 100 µm, puis une seconde étape sur un tamis de 40 µm.After neutralization of the enzymatic activity, by adding, for example, fetal calf serum, autologous human serum, or allologous human serum from a compatible group, or an enzymatic activity inhibitor, the product digestion is then filtered through sieves, under gravity pressure to remove undissociated fibers and collect the cells detached from the muscle fibers. Preferably, a filtration step will be carried out in two steps: a filtration step on a 100 μm sieve, then a second step on a 40 μm sieve.

Les cellules recueillies après filtration sont transvasées dans un réacteur de culture en présence d'un milieu dont la composition permet leur croissance ou/et leur différenciation. La composition du milieu est choisie en fonction du type cellulaire dominant souhaité en fin de culture. A ce stade, une partie de la préparation peut être congelée. Cela peut être une partie de la préparation initiale ou une sous-population issue d'une étape d'enrichissement ou de déplétion. Les milieux de culture utilisés contiennent un ou plusieurs facteurs de croissance et/ou de différenciation dont le rôle est de diriger les cellules progénitrices vers une voie de différenciation spécifique et de les faire proliférer. A titre d'exemple de facteurs de croissance, on peut citer les facteurs de croissance des fibroblastes, bFGF, aFGF, FGF6, le facteur de croissance des hépatocytes, HGF/SF, de l'épiderme, EGF et les différents facteurs caractérisés tels IGF-1, PDGF, LIF, VEGF, SCF, TGFb, TNFa, IL-6, IL-15, NGF, la neuréguline, la thrombopoïétine et l'hormone de croissance. On peut les associer à différentes hormones ou molécules actives qui peuvent entrer dans la composition des milieux, telles que les glucocorticoïdes (naturels ou hémi-synthétiques, i.e. l'hydrocortisone, la dexaméthasone, la prednisolone ou la triamcinolone), les progéstagènes et dérivés (progestérone), les oestrogènes et dérivés (oestradiol), les androgène et dérivés (testostérone), les minéralocarticoïdes et dérivés (aldostérone), les hormones LH, LH-RH, FSH et TSH, les hormones thyroïdiennes T3, T4, l'acide rétinoïque et ses dérivés, la calcitonine, les prostaglandines E2 et F2/alpha ou l'hormone parathyroïdienne.The cells collected after filtration are transferred to a culture reactor in the presence of a medium whose composition allows their growth and / or their differentiation. The composition of the medium is chosen according to the dominant cell type desired at the end of culture. At this stage, some of the preparation can be frozen. This can be a part of the initial preparation or a subpopulation from an enrichment or depletion stage. The culture media used contain one or more growth and / or differentiation factors whose role is to direct the progenitor cells to a specific differentiation pathway and to proliferate them. By way of example of growth factors, mention may be made of fibroblast growth factors, bFGF, aFGF, FGF6, hepatocyte growth factor, HGF / SF, epidermal factor, EGF and the various factors characterized such as IGFs. -1, PDGF, LIF, VEGF, SCF, TGFb, TNFa, IL-6, IL-15, NGF, neuregulin, thrombopoietin and growth hormone. They can be associated with different hormones or active molecules that can be used in the composition of media, such as glucocorticoids (natural or semi-synthetic, ie hydrocortisone, dexamethasone, prednisolone or triamcinolone), progestagens and derivatives ( progesterone), estrogens and derivatives (estradiol), androgens and derivatives (testosterone), mineralocarticoids and derivatives (aldosterone), LH hormones, LH-RH, FSH and TSH, thyroid hormones T3, T4, retinoic acid and its derivatives, calcitonin, prostaglandins E2 and F2 / alpha or parathyroid hormone.

Avant la mise en culture, ou au cours d'une phase d'expansion, la préparation peut faire l'objet d'enrichissement ou de déplétions. Ces opérations sont réalisées par l'homme du métier en utilisant les différentes techniques proposées dans l'état de la technique pour opérer un tri sélectif. Ces techniques reposent sur l'identification d'antigènes extracellulaires caractéristiques de tel ou tel type cellulaire par des réactifs spécifiques. A titre d'exemple, les antigènes CD34 et CD56 exprimés sur certaines cellules présentes au sein de la population de cellules musculaires peuvent être utilisés. Le tri Initial de la population cellulaire totale selon l'expression de ces deux antigènes permet ainsi de séparer deux groupes. En particulier, il permet de séparer les types cellulaires CD34+ des types cellulaires CD34. Il a été montré que la population CD34+ génère un type cellulaire dominant CD15+, CD56- n'exprimant pas la desmine. La population CD34 génère un type cellulaire dominant CD56+, CD15-exprimant la desmine. Dans de rares cas, la population CD34- génère une population CD56-, CD15-, En particulier, le procédé de l'invention comprend une étape de déplétion des cellules CD34- ou CD34+ conduisant respectivement à un population comprenant un type cellulaire dominant C015+ ou CD56+.Prior to culturing, or during an expansion phase, the preparation may be enriched or depleted. These operations are carried out by those skilled in the art using the various techniques proposed in the state of the art to operate a selective sorting. These techniques rely on the identification of extracellular antigens characteristic of this or that cell type by specific reagents. By way of example, CD34 and CD56 antigens expressed on certain cells present within the muscle cell population may be used. Initial sorting of the total cell population according to the expression of these two antigens thus makes it possible to separate two groups. In particular, it makes it possible to separate CD34 + cell types from CD34 cell types. It has been shown that the CD34 + population generates a dominant CD15 + cell type, CD56- not expressing desmin. The CD34 population generates a dominant CD56 +, CD15-expressing desmin cell type. In rare cases, the population CD34- generates a population CD56-, CD15-. In particular, the method of the invention comprises a stage of depletion of CD34- or CD34 + cells leading respectively to a population comprising a dominant cell type C015 + or CD56 +.

Un mode de réalisation du procédé de l'invention porte donc sur un procédé d'obtention d'une population de cellules dont un type cellulaire dominant est le type cellulaire myoblastique caractérisé en ce qu'il comprend une étape de déplétion des cellules CD34+ avant la mise en culture des cellules ou au cours de l'une des phases d'expansion.One embodiment of the method of the invention therefore relates to a method for obtaining a population of cells of which a dominant cell type is the myoblastic cell type, characterized in that it comprises a stage of depletion of CD34 + cells before the cell culture or during one of the expansion phases.

Les cellules sont ensuite mises en culture dans un réacteur adapté pour la culture de cellules adhérentes. Afin de s'affranchir des contraintes de contrôles de la vitesse d'agitation, de sa régularité et de l'homogénéité des préparations, le réacteur de culture est de préférence statique. Il doit présenter une grande surface de culture par rapport aux supports classiques (boites de Pétri, flasques) de manière à récolter en quelques jours une population cellulaire importante. Une exemple d'un tel réacteur de culture est le dispositif de culture en plateaux (simple, double et/ou multi-étagé).The cells are then cultured in a reactor suitable for the culture of adherent cells. In order to overcome the constraints of controlling the stirring speed, its regularity and the homogeneity of the preparations, the culture reactor is preferably static. It must have a large culture surface compared to conventional carriers (petri dishes, flasks) so as to harvest in a few days a large cell population. An example of such a culture reactor is the tray culture device (single, double and / or multi-stage).

Le dispositif de culture utilisé dans le procédé permet de plus l'échantillonnage des cellules de manière stérile afin de réaliser les prélèvements nécessaires à l'identification des types cellulaires présents aux différents stades de la culture par analyse de marqueurs spécifiques. Il permet la vidange des milieux, le lavage et le décollement des cellules et enfin leur récolte de manière stérile.The culture device used in the method also allows sterile sampling of the cells in order to carry out the samples necessary for the identification of the cell types present at the different stages of the culture by analysis of specific markers. It allows the emptying of media, the washing and detachment of cells and finally their harvest in a sterile manner.

De manière préférée, des poches sont utilisées et des tubulures stériles spécialement adaptées relient les poches au réacteur pour permettre les transvasements des milieux ou la récolte des cellules. Ce dispositif permet ainsi de réaliser un grand nombre d'opérations en système clos. Selon la population cellulaire souhaitée, le nombre de jours de culture varie de 0 à 45 jours.In a preferred manner, bags are used and specially adapted sterile tubings connect the bags to the reactor to allow transfer of media or cell harvesting. This device thus makes it possible to perform a large number of operations in a closed system. Depending on the desired cell population, the number of days of culture varies from 0 to 45 days.

De plus, la culture peut être poursuivie par des techniques classiques d'expansion ou de perfusion pendant une durée pouvant atteindre plusieurs mois.In addition, the culture can be continued by conventional expansion or perfusion techniques for a period of up to several months.

Afin d'augmenter le nombre de cellules récoltées, une ou plusieurs phases d'expansion sont possibles. Les phases d'expansion comprennent une étape de décollement des cellules, de lavage des cellules et de remise en culture sur une plus grande surface de culture, les solutions et enzymes utilisées pour réaliser ces étapes étant bien connues de l'homme du métier. En particulier, dans un mode de réalisation préféré utilisant un milieu de culture approprié pour la différentiation en myoblaste, le procédé de l'invention comprend au moins trois phases d'expansions de cellules. Un tel procédé permet de multiplier le nombre de cellules sans modifier sensiblement les proportions des types cellulaires obtenues en fin de culture de chaque expansion.In order to increase the number of cells harvested, one or more expansion phases are possible. The expansion phases comprise a step of detaching the cells, washing the cells and restoring culture to a larger culture surface, the solutions and enzymes used to carry out these steps being well known to those skilled in the art. In particular, in a preferred embodiment using a culture medium suitable for myoblast differentiation, the method of the invention comprises at least three phases of cell expansions. Such a method makes it possible to multiply the number of cells without substantially modifying the proportions of the cell types obtained at the end of culture of each expansion.

Une cinétique de différenciation est réalisée dans le procédé de l'invention par l'identification des types cellulaires présents dans les populations de cellules obtenues aux différents stades de la culture. Dans le texte qui suit, on désignera l'étape d'identification des types cellulaires présents aux différents stades de la culture par le terme « caractérisation des populations cellulaires ». Cette caractérisation est réalisée à partir d'échantillons prélevés lors de la mise en culture, au cours de la culture et lors de la récolte des cellules. La caractérisation des populations cellulaires peut aussi porter sur une culture de cellules réalisée en parallèle à plus petite échelle mais dans des conditions identiques ou équivalentes en terme de milieu de culture et de procédé de réalisation des phases d'expansion. La caractérisation porte sur les cellules adhérentes ou les cellules présentes dans le surnageant. Les différents stades de la culture sont comptés en jours à partir du jour de mise en culture en réacteur jusqu'à l'apparition des premières cellules différenciées ou l'obtention d'un degré de confluence des cellules d'au moins 20 à 50%. La caractérisation des populations de cellules a pour fonction l'identification de l'ensemble des types cellulaires et de leurs proportions en fonction du stade de la culture. Elle permet en particulier de mettre en évidence les types cellulaires dominants en fonction du stade de la culture. Le procédé de l'invention propose ainsi un moyen de choisir une population cellulaire en fonction des caractéristiques des différents types cellulaires présents dans la population.Kinetics of differentiation is carried out in the method of the invention by identifying cell types present in the cell populations obtained at different stages of the culture. In the text that follows, we will designate the step of identifying the cell types present at different stages of the culture by the term "characterization of cell populations". This characterization is carried out from samples taken during the cultivation, during the culture and during the harvesting of the cells. The characterization of the cell populations can also relate to a cell culture carried out in parallel on a smaller scale but under identical or equivalent conditions in terms of culture medium and method of carrying out the expansion phases. The characterization relates to the adherent cells or the cells present in the supernatant. The different stages of the culture are counted in days starting from the day of culture in a reactor until the appearance of the first differentiated cells or obtaining a degree of confluence of the cells of at least 20 to 50%. . The function of cell population characterization is to identify all cell types and their proportions according to the stage of culture. In particular, it makes it possible to highlight the dominant cell types according to the stage of the culture. The method of the invention thus provides a means of selecting a cell population according to the characteristics of the different cell types present in the population.

Cette caractérisation est réalisée par l'analyse de marqueurs cellulaires par cytofluorimétrie de flux ou FACS, après marquage des antigènes de surfaces ou de tout antigène spécifique des différents types cellulaires à analyser. Dans le présent texte, le terme « marqueurs cellulaires » indique tout antigène cellulaire permettant d'apporter des informations à lui seul ou en combinaison avec d'autres marqueurs sur un type cellulaire. Tout type de marqueur cellulaire peut être utilisé par le procédé, le choix des marqueurs utilisés sera guidé par le choix des types cellulaires que l'on souhaite caractériser.This characterization is carried out by the analysis of cell markers by flow cytofluorometry or FACS, after labeling the surface antigens or any specific antigen of the different cell types to be analyzed. In the present text, the term "cellular markers" denotes any cell antigen that can provide information alone or in combination with other markers on a cell type. Any type of cell marker can be used by the method, the choice of markers used will be guided by the choice of cell types that one wishes to characterize.

Toute technique apte à caractériser un antigène cellulaire peut être mise en oeuvre dans le procédé de l'invention. On peut citer à titre d'exemple sans être limitatif, le Western Blot ou l'immunocytochimie. Les techniques permettant la caractérisation des ARN messagers codant lesdits antigènes sont utilisables de manière équivalente.Any technique capable of characterizing a cellular antigen can be implemented in the method of the invention. By way of example, Western Blot or immunocytochemistry may be mentioned without being limiting. The techniques allowing the characterization of messenger RNAs encoding said antigens can be used in an equivalent manner.

Les marqueurs cellulaires analysés pour l'identification des types cellulaires sont spécifiquement choisis parmi les marqueurs suivants : CD10, CD13, CD15, CD16, CD34, CD38, CD40, CD44, CD45, CD56, CD71, CD80, CD117 ou encore les structures suivantes, CD138, HLA-Classe I, HLA-classe II, VLA3, VLA5, VLA6, ICAM-1, VCAM et la desmine. De manière préférée, les marqueurs CD10, CD13, CD15, CD34, CD44, CD56, CD117 et HLA Classe I et la desmine sont utilisés. Ces marqueurs sont identifiés par l'utilisation d'anticorps monoclonaux spécifiques. Le tableau 1, ci-après fourni dans la partie expérimentale, indique, pour chacun des marqueurs, les types cellulaires exprimant classiquement les antigènes correspondants. Il est à noter que ces marqueurs cellulaires ont été développés initialement pour la caractérisation du système immuno-hématologique. Une originalité de l'invention consiste en l'utilisation de ces marqueurs pour la caractérisation des cellules d'origine musculaire aux différentes étapes de la culture ou de l'expansion.The cellular markers analyzed for the identification of cell types are specifically chosen from the following markers: CD10, CD13, CD15, CD16, CD34, CD38, CD40, CD44, CD45, CD56, CD71, CD80, CD117 or the following structures, CD138, HLA-Class I, HLA-class II, VLA3, VLA5, VLA6, ICAM-1, VCAM and desmin. In a preferred manner, the markers CD10, CD13, CD15, CD34, CD44, CD56, CD117 and HLA Class I and desmin are used. These markers are identified by the use of specific monoclonal antibodies. Table 1, hereinafter provided in the experimental part, indicates, for each of the markers, the cell types which conventionally express the corresponding antigens. It should be noted that these cell markers were initially developed for the characterization of the immuno-hematological system. An originality of the invention consists in the use of these markers for the characterization of cells of muscular origin at the different stages of culture or expansion.

Le procédé de l'invention est donc mis en oeuvre grâce à l'analyse de marqueurs cellulaires non classiquement utilisés dans la caractérisation des cellules issues de tissus musculaires. Ces marqueurs cellulaires sont par exemple CD10, CD13, CD15, CD34, CD44, CD45, CD117 et HLA Classe I.The method of the invention is therefore implemented through the analysis of cellular markers not conventionally used in the characterization of cells derived from muscle tissue. These cell markers are, for example, CD10, CD13, CD15, CD34, CD44, CD45, CD117 and HLA Class I.

Il a été effectivement observé que la mise en oeuvre du procédé de l'invention permet la mise en évidence d'une évolution au cours du temps du type cellulaire dominant présent dans la culture. De manière surprenante, aux stades précoces de la mise en culture des cellules de tissus musculaires et dans un milieu adapté pour la différenciation myoblastique, les populations majoritaires sont celles exprimant les marqueurs de cellules progénitrices de la moelle et des cellules du système lymphoblastoïde. Plus précisément, il a été observé à J0 et J1, une majorité de cellules de phénotype CD34+/45- ainsi que la présence d'un type cellulaire minoritaire CD117+ dans la population de cellules non adhérentes. L'évolution est ensuite marquée par une augmentation croissante de la proportion de cellules CD15+ et/ou CD56+, et une diminution des populations CD34+. Néanmoins, le type cellulaire dominant reste CD34+ de J0 à J5. Une transition progressive d'une population de type cellulaire dominant CD34+ à CD56+ est observée. Par ailleurs, la proportion des types cellulaires CD13+, CD44+, CD71+ augmente avec le temps. Des populations minoritaires CD138+, HLA-Classe II+ et CD38+ sont aussi observées. Au terme de la culture, le type cellulaire dominant est de phénotype CD56+, en particulier à partir du stade J5 jusqu'à la fin de la culture. Les cellules CD56+ expriment également CD10, CD13, CD44, la desmine, HLA-Classe I, CD44, VLA-3, VLA-5 et VLA-6. Ces marqueurs caractérisent les cellules myoblastiques. Un deuxième type cellulaire prépondérant est constitué de cellules exprimant CD15, CD10, CD13 et HLA-Classe I. Une population CD56-/CD15-/CD13+ est aussi présente en proportions variables constituant un troisième type cellulaire prépondérant. Dans cette population, une partie exprime la desmine et une partie ne l'exprime pas. La mise en oeuvre du procédé de l'invention a ainsi permis de distinguer trois types cellulaires présents dans les populations de cellules, dont les proportions respectives évoluent de manière importante au cours de la culture, les cellules CD34+, les cellules CD15+ et les cellules CD56+.It has indeed been observed that the implementation of the method of the invention allows the demonstration of an evolution over time of the dominant cell type present in the culture. Surprisingly, in the early stages of culturing muscle tissue cells and in a medium suitable for myoblastic differentiation, the majority populations are those expressing markers of marrow progenitor cells and cells of the lymphoblastoid system. More precisely, a majority of CD34 + / 45- phenotype cells were observed on day 0 and day 1, as well as the presence of a CD117 + minority cell type in the non-adherent cell population. The evolution is then marked by an increasing increase in the proportion of CD15 + and / or CD56 + cells, and a decrease in CD34 + populations. Nevertheless, the dominant cell type remains CD34 + from D0 to D5. A gradual transition from a dominant cell type CD34 + to CD56 + cell is observed. On the other hand, the proportion of CD13 +, CD44 +, CD71 + cell types increases with time. Minority populations CD138 +, HLA-Class II + and CD38 + are also observed. At the end of the culture, the dominant cell type is of CD56 + phenotype, especially from the J5 stage until the end of the culture. CD56 + cells also express CD10, CD13, CD44, desmin, HLA-class I, CD44, VLA-3, VLA-5 and VLA-6. These markers characterize myoblastic cells. A second predominant cell type consists of cells expressing CD15, CD10, CD13 and HLA-Class I. A population CD56- / CD15- / CD13 + is also present in variable proportions constituting a third predominant cell type. In this population, one part expresses the desmin and one part does not express it. The implementation of the method of the invention has thus made it possible to distinguish three cell types present in the cell populations, the respective proportions of which change significantly during the course of the culture, the CD34 + cells, the CD15 + cells and the CD56 + cells. .

Dans un mode de mise en oeuvre préféré du procédé de l'invention, la caractérisation des populations cellulaires consiste à déterminer le pourcentage respectif des trois types cellulaires CD34+, CD15+ et CD56+ aux différentes étapes de la culture.In a preferred embodiment of the method of the invention, the characterization of the cell populations consists in determining the respective percentage of the three CD34 +, CD15 + and CD56 + cell types at the different stages of the culture.

Dans une forme de réalisation de l'invention, un type cellulaire recherché dans une population cellulaire est séparé après sélection du stade de culture pendant lequel le type cellulaire recherché est dans l'état qualitatif ou quantitatif recherché, en particulier, après sélection du stade de culture pendant lequel le type cellulaire recherché est dominant. La présence en proportion dominante des cellules à séparer facilite leur préparation. L'invention fournit ainsi un procédé d'obtention d'une population cellulaire de degré de pureté élevé. On entend par population cellulaire de degré de pureté élevé, une population de cellules dans laquelle le type cellulaire dominant est en proportion supérieure à 50% dans la population cellulaire. Il apparaît clairement que l'homme du métier pourra mettre en oeuvre les différentes techniques proposées dans l'état de l'art pour faire un tri sélectif des dites cellules. A titre d'exemple, citons les techniques de tri par clonage, par cytofluorimétrie de flux ou encore par colonnes d'immunoaffinité ou immunomagnétiques utilisant des anticorps spécifiques des cellules en cause.In one embodiment of the invention, a desired cell type in a cell population is separated after selection of the culture stage during which the desired cell type is in the desired qualitative or quantitative state, particularly after selection of the culture during which the desired cell type is dominant. The presence in predominant proportion of the cells to be separated facilitates their preparation. The invention thus provides a method for obtaining a cell population of high purity. By high purity cell population is meant a population of cells in which the dominant cell type is in excess of 50% in the cell population. It is clear that one skilled in the art can implement the various techniques proposed in the state of the art for a selective sorting of said cells. By way of example, mention may be made of cloning, flow cytofluorometry or immunoaffinity or immunomagnetic columns using antibodies specific for the cells in question.

Dans un autre mode de réalisation, au contraire, le procédé de l'invention est caractérisé en ce qu'il ne comprend pas d'étapes de purification, de sélection positive, ou de clonage d'un type cellulaire spécifique. Par sélection positive, il faut comprendre toute étape permettant de trier les cellules sur la base d'au moins une caractéristique particulière et notamment l'expression d'un marqueur cellulaire. Il a été montré en particulier, que le procédé de l'invention, contrairement aux procédés décrits dans l'état de la technique, permet d'obtenir un produit de thérapie cellulaire comprenant un type cellulaire dominant de type myoblaste, sans étape de sélection préférentielle du type cellulaire myoblastique. L'absence d'étapes de clonage, de purification ou de sélection positive, permet d'améliorer nettement le rendement final obtenu au terme des procédures d'expansion en terme de nombres de cellules obtenues.In another embodiment, on the contrary, the method of the invention is characterized in that it does not include purification steps, positive selection, or cloning of a specific cell type. By positive selection, it is necessary to understand any step for sorting the cells on the basis of at least one particular characteristic and in particular the expression of a cellular marker. It has been shown in particular that the method of the invention, unlike the methods described in the state of the art, makes it possible to obtain a cell therapy product comprising a dominant myoblast-type cell type, without a preferential selection step. of the myoblastic cell type. The absence of cloning steps, purification or positive selection, significantly improves the final yield obtained after the expansion procedures in terms of numbers of cells obtained.

Dans le procédé de l'invention, après arrêt de la culture au stade de culture choisi, un aliquot de cellules peut être prélevé et remis en culture séparément. Le milieu peut être le même ou de composition différente. Les milieux de culture sont choisis en fonction de la voie de différenciation souhaitée. Un exemple de milieu permettant la différenciation en myoblastes, en cellules endothéliales, en cellules musculaires lisses ou en myofibroblastes est le milieu MCDB 120 supplémenté par du sérum de veau foetal (23) qui comprend en particulier un glucocorticoïde tel que la dexaméthasone et le bFGF. Un autre exemple de milieu préféré est le milieu MCDB 120 modifié en substitutant la L-valine par la D-valine. Il a en effet été constaté qu'une telle modification permet d'obtenir un milieu particulièrement sélectif pour la différenciation des cellules en myoblastes. Un tel milieu permet d'obtenir, avec le procédé de l'invention une population comprenant un très grand nombre de cellules dont une majorité est de type CD56+ ou HLA Classe 1.In the method of the invention, after stopping the culture at the selected culture stage, an aliquot of cells can be removed and cultured separately. The medium may be the same or of different composition. The culture media are selected according to the desired differentiation pathway. An example of a medium for differentiation into myoblasts, endothelial cells, smooth muscle cells or myofibroblasts is the MCDB 120 medium supplemented with fetal calf serum (23) which in particular comprises a glucocorticoid such as dexamethasone and bFGF. Another example of a preferred medium is the modified MCDB 120 medium substituting L-valine for D-valine. It has indeed been found that such a modification makes it possible to obtain a particularly selective medium for the differentiation of the cells into myoblasts. Such a medium makes it possible to obtain, with the method of the invention, a population comprising a very large number of cells, most of which are of the CD56 + or HLA Class 1 type.

Un milieu de différenciation en tissus adipeux contient en particulier la dexaméthasone, l'isobutyl-methylxanthine, et le cas échéant, du sérum de veau foetal, l'indométhacine et l'insuline. Le maintien de la différenciation est obtenu dans un milieu contenant 10% de sérum de veau foetal et l'insuline. Pour obtenir une différenciation en tissus cartilagineux, les cellules sont centrifugées en micromasses et cultivées dans du milieu sans sérum mais contenant du TGF-béta3. Un milieu de différenciation en tissus osseux contient de la dexaméthasone, du béta-glycérolphosphate, de l'ascorbate, et le cas échéant, du sérum de veau foetal.A differentiation medium in adipose tissue contains, in particular, dexamethasone, isobutylmethylxanthine and, if appropriate, serum fetal calf, indomethacin and insulin. Maintaining differentiation is achieved in medium containing 10% fetal calf serum and insulin. To obtain a differentiation in cartilage tissue, the cells are centrifuged in micromass and cultured in medium without serum but containing TGF-beta3. A bone tissue differentiation medium contains dexamethasone, beta-glycerol phosphate, ascorbate, and, where appropriate, fetal calf serum.

La récolte des cellules s'effectue au stade de culture choisi en fonction de la population cellulaire dont on souhaite l'obtention. Les cellules non adhérentes présentes dans le surnageant ou/et les cellules adhérentes peuvent être récoltées par ce procédé. Les populations de cellules présentes dans le surnageant et de cellules adhérentes peuvent être de composition différente. Par conséquent, le mode de récolte des cellules sera fonction de la population cellulaire recherchée. La récolte des cellules adhérentes s'effectue par dissociation enzymatique des cellules et décollement de la couche cellulaire par des techniques connues de l'homme de métier. La récolte des cellules non adhérentes se fait par aspiration.The cells are harvested at the selected culture stage depending on the cell population that is desired. Non-adherent cells present in the supernatant and / or adherent cells can be harvested by this method. The cell populations present in the supernatant and adherent cells may be of different composition. Consequently, the mode of harvesting the cells will be a function of the desired cell population. The harvesting of the adherent cells is carried out by enzymatic dissociation of the cells and detachment of the cell layer by techniques known to those skilled in the art. The non-adherent cells are harvested by aspiration.

La mise en oeuvre du procédé ci-dessus incluant l'établissement d'une cinétique de différenciation à partir de cellules issues de biopsies de tissus musculaires permet l'obtention de populations cellulaires caractérisées.The implementation of the above method including the establishment of kinetics of differentiation from cells derived from muscle tissue biopsies allows obtaining characterized cell populations.

Les populations de cellules peuvent être obtenues par un procédé similaire au procédé d'obtention de populations de cellules décrit plus haut, mais ne comprenant pas d'étape d'identification des types cellulaires aux différents stades cellulaires. En effet, l'étape d'identification des types cellulaires est nécessaire pour le choix du stade de récolte. Dès lors que, ayant mis en oeuvre au moins une fois le procédé de l'invention, l'homme du métier connaît le stade optimal de récolte des cellules en fonction de la population recherchée, celui-ci peut obtenir les mêmes types de populations en limitant le nombre de marqueurs utilisés pour la caractérisation des populations de cellules et les stades au cours desquels il effectue cette caractérisation. Ainsi, par « populations de cellules susceptibles d'être obtenues », il faut donc inclure une population de cellules obtenue par le procédé de l'invention ne comprenant pas nécessairement une étape de caractérisation des populations de cellules au sens de l'invention, telle que décrite plus haut. Le stade de récolte optimal étant connu par une mise en oeuvre d'une cinétique de différenciation sur une culture préalable, effectuée dans les mêmes conditions.The cell populations can be obtained by a method similar to the method for obtaining cell populations described above, but not including a step of identifying the cell types at different levels. cellular stages. Indeed, the step of identifying the cell types is necessary for the choice of the harvesting stage. Since, having carried out at least once the process of the invention, the skilled person knows the optimal stage of harvesting cells according to the desired population, it can obtain the same types of populations by limiting the number of markers used for the characterization of cell populations and the stages at which it performs this characterization. Thus, by "populations of cells that can be obtained", it is therefore necessary to include a population of cells obtained by the process of the invention not necessarily comprising a step of characterizing cell populations within the meaning of the invention, such as as described above. The optimal harvest stage is known by implementing a differentiation kinetics on a prior culture, carried out under the same conditions.

Les différentes populations cellulaires susceptibles d'être obtenues par le procédé de l'invention, avec ou sans étape d'identification sont de différents types selon le stade de culture choisi.The different cell populations that can be obtained by the method of the invention, with or without an identification step, are of different types depending on the culture stage chosen.

En particulier, aux stades précoces de culture des cellules, l'invention fournit une population cellulaire dont le type cellulaire dominant exprime le marqueur CD34. Lorsque seules les cellules non-adhérentes sont récoltées au stade précoce de culture, la population cellulaire comprend aussi un type cellulaire minoritaire de phénotype CD117+.In particular, at the early stages of cell culture, the invention provides a cell population whose dominant cell type expresses the CD34 marker. When only the non-adherent cells are harvested at the early stage of culture, the cell population also includes a minority cell type of CD117 + phenotype.

Par la mise en oeuvre du procédé décrit ci-dessus dans lequel le milieu de culture est adapté pour la différenciation myoblastique, l'invention fournit une population cellulaire dont le type cellulaire dominant exprime le marqueur CD15.By carrying out the method described above in which the culture medium is adapted for myoblastic differentiation, the invention provides a cell population whose dominant cell type expresses the CD15 marker.

Enfin, par la mise en oeuvre du procédé décrit ci-dessus et en utilisant un milieu de culture adapté pour la différenciation myoblastique, une forme de réalisation préférée de l'invention fournit une population cellulaire dont le type cellulaire dominant est constitué de cellules myoblastiques. Les cellules myoblastiques sont caractérisées par l'analyse de l'expression du marqueur CD56. Elles sont caractérisées de manière préférée par l'analyse de l'expression du marqueur CD56 en combinaison avec l'analyse des marqueurs CD10, CD13, CD44, HLA Classe I et la desmine. L'analyse de l'expression de' la desmine, protéine intracellulaire du cytosquelette spécifique des cellules myoblastiques et des cellules musculaires différenciées nécessite un protocole adapté de marquage et d'analyse par FACS, détaillé dans la partie expérimentale. La population cellulaire comprend en plus une population doublement négative CD56- /CD15-.Finally, by carrying out the method described above and using a culture medium suitable for myoblastic differentiation, a preferred embodiment of the invention provides a cell population whose dominant cell type is myoblastic cells. Myoblastic cells are characterized by the analysis of CD56 marker expression. They are preferably characterized by CD56 marker expression analysis in combination with the analysis of CD10, CD13, CD44, HLA Class I and desmin markers. Analysis of the expression of desmin, an intracellular cytoskeletal protein specific to myoblastic cells and differentiated muscle cells requires a suitable FACS labeling and analysis protocol, detailed in the experimental section. The cell population additionally comprises a doubly negative population CD56 - / CD15 - .

L'invention fournit en particulier une population cellulaire issue d'une même biopsie musculaire et comprenant de 50 x 106 à 800 x 109 cellules, de préférence au moins 500 x 106 cellules dont au moins 50% des cellules, et mieux au moins 60%, et de manière encore préférée, au moins 70%, sont CD56+.The invention provides in particular a cell population derived from the same muscle biopsy and comprising from 50 × 10 6 to 800 × 10 9 cells, preferably at least 500 × 10 6 cells, of which at least 50% of the cells, and better still at least 60%, and more preferably at least 70%, are CD56 +.

L'invention porte sur l'utilisation d'une population de cellules CD56+ obtenue selon les procédés décrits plus haut dans la préparation d'un produit de thérapie cellulaire pour le traitement chez l'homme de l'insuffisance cardiaque post-ischémique ou pour la réparation des tissus cardiaques. En effet, la mise en oeuvre du procédé permet l'obtention d'un grand nombre de cellules rapidement et les populations cellulaires obtenues ont l'avantage d'être homogènes, et donc particulièrement adaptées pour la préparation d'un produit de thérapie cellulaire.The invention relates to the use of a population of CD56 + cells obtained according to the methods described above in the preparation of a cellular therapy product for the treatment in humans of post-ischemic heart failure or for repair of heart tissue. Indeed, the implementation of the method makes it possible to obtain a large number of cells rapidly and the cell populations obtained have the advantage of being homogeneous, and therefore particularly suitable for the preparation of a cell therapy product.

Elle porte aussi sur l'utilisation d'une population de cellules issues d'une même biopsie musculaire et comprenant de 500x106 à 800 x 109 cellules dont au moins 50%, et mieux au moins 60%, sont CD56+ ou HLA Classe 1 dans la préparation d'un produit de thérapie cellulaire pour le traitement chez l'homme, de l'insuffisance cardiaque post-ischémique ou toutes cardiopathies d'origine génétique, virale, médicamenteuse, infectieuse ou parasitaireIt also relates to the use of a population of cells from the same muscle biopsy and comprising from 500x10 6 to 800 x 10 9 cells of which at least 50%, and better still at least 60%, are CD56 + or HLA Class 1 in the preparation of a cellular therapy product for treatment in humans, post-ischemic heart failure or any genetic, viral, medicinal, infectious or parasitic heart disease

Le traitement des cardiopathies consiste en particulier à injecter à l'aide d'une aiguille une population de cellules, dont le type dominant a les caractéristiques de cellules myoblastiques, obtenue et préparée comme produit de thérapie cellulaire, directement dans le tissu myocardique (12) ou indirectement dans la circulation artérielle (24).The treatment of heart disease consists in particular of injecting with a needle a population of cells, the dominant type has the characteristics of myoblastic cells, obtained and prepared as a cell therapy product, directly in the myocardial tissue (12) or indirectly in the arterial circulation (24).

De préférence, au moins 600 x 106 cellules issues d'une même biopsie sont injectées.Preferably, at least 600 x 10 6 cells from the same biopsy are injected.

L'insuffisance cardiaque est aujourd'hui prise en charge par traitement par des inhibiteurs de l'enzyme de conversion de l'angiotensine (ACEI). Les expériences décrites ci-après indiquent un effet améliorant certain de la transplantation de cellules myoblastiques in situ dans la zone infarcie lorsque cette transplantation est réalisée concomitamment au traitement par l'ACEI.Heart failure is now managed by treatment with angiotensin converting enzyme (ACEI) inhibitors. The experiments described below indicate a definite improvement effect of myoblastic cell transplantation in situ in the infarcted zone when this transplantation is performed concomitantly with CIRA treatment.

L'invention porte donc également sur l'utilisation de populations cellulaires d'origine musculaire obtenues selon les procédés décrits plus haut comme transplant pour potentialiser les traitements pharmacologiques des insuffisances cardiaques.The invention therefore also relates to the use of cell populations of muscle origin obtained according to the methods described above as a transplant for potentiating the pharmacological treatments of heart failure.

La partie expérimentale du présent texte décrit la réalisation de transplantations de cellules d'origine musculaire autologues chez le rat démontrant la faisabilité de cette technique. En effet, les résultats montrent que la transplantation de ces cellules chez le rat améliore significativement les paramètres d'évaluation fonctionnelle démontrant par là la faisabilité de telles transplantations. Ils montrent aussi que l'amélioration de la fonction myocardique est liée au nombre de cellules greffées.The experimental part of the present text describes the carrying out of autologous muscle cell transplantations in rats demonstrating the feasibility of this technique. Indeed, the results show that the transplantation of these cells in the rat significantly improves the functional evaluation parameters thus demonstrating the feasibility of such transplants. They also show that the improvement in myocardial function is related to the number of grafted cells.

La partie expérimentale décrit également les résultats obtenus chez le rat, lors de l'utilisation conjointe de cellules musculaires autologues et du traitement pharmacologique de l'insuffisance cardiaque.The experimental part also describes the results obtained in the rat, during the joint use of autologous muscle cells and the pharmacological treatment of heart failure.

Au cours des phases de culture et d'expansion des cellules dans les procédés fournis par l'invention, une étape de modification génétique des cellules par transfection d'un acide nucléique hétérologue peut être réalisée. L'acide nucléique est choisi de manière à permettre l'expression d'un polypeptide ou d'une protéine dans les cellules transfectées. Les cellules transfectées sont ensuite transplantées et permettent la délivrance du polypeptide ou de la protéine exprimés à partir de l'acide nucléique hétérologue, le dit polypeptide ou protéine étant un produit biologiquement actif. L'invention porte ainsi sur l'utilisation d'une population de cellules comme produit de thérapie cellulaire en tant que plate-forme de délivrance d'un produit biologiquement actif.During the cell growth and expansion phases in the methods provided by the invention, a step of genetic modification of the cells by transfection of a heterologous nucleic acid can be performed. The nucleic acid is selected to allow expression of a polypeptide or protein in the transfected cells. The transfected cells are then transplanted and allow delivery of the polypeptide or protein expressed from the heterologous nucleic acid, wherein said polypeptide or protein is a biologically active product. The invention thus relates to the use of a cell population as a cell therapy product as a delivery platform for a biologically active product.

La première partie présente les exemples de mise en oeuvre du procédé permettant selon le stade de culture choisi l'obtention de populations cellulaires dont le type cellulaire dominant est CD34+ ou CD15+ ou CD56+ (myoblastique) ou doublement négatif CD56-/CD15-.The first part presents the examples of implementation of the method according to the selected culture stage for obtaining cell populations whose dominant cell type is CD34 + or CD15 + or CD56 + (myoblastic) or double negative CD56- / CD15-.

Les résultats présentés en deuxième partie montrent l'efficacité de la technique de transplantation de cellules myoblastiques sur des tissus cardiaques infarcis chez le rat. Elle permet aussi de déterminer les critères requis pour une bonne efficacité.The results presented in the second part show the efficiency of the technique of transplantation of myoblastic cells on tissues cardiac infarction in the rat. It also makes it possible to determine the criteria required for a good efficiency.

Enfin, une troisième partie présente les essais cliniques réalisés chez l'homme de transplantation de cellules d'origine musculaire pour la reconstitution de tissus myocardiques, la réparation de tissus myocardiques, la génération de tissu métaboliquement actif, la génération de tissu présentant une activité fonctionnelle n'existant pas avant reconstitution. Elle montre en outre que les cellules musculaires peuvent également jouer un rôle dans le remodelage du tissu cardiaque chez l'homme.Finally, a third part presents human clinical trials of muscle cell transplantation for myocardial tissue reconstitution, myocardial tissue repair, metabolically active tissue generation, tissue generation with functional activity. does not exist before reconstitution. It also shows that muscle cells can also play a role in remodeling heart tissue in humans.

PARTIE EXPERIMENTALEEXPERIMENTAL PART Légende des figures:Legend of figures:

Figure 1 : Figure 1 :
Graphe indiquant les valeurs de LVEF entre le groupe traité et le groupe contrôle.Graph showing the LVEF values between the processed group and the control group.
Figure 2 : Figure 2 :
Graphe indiquant les valeurs de LVEDV entre le groupe traité et le groupe contrôle.Graph showing the LVEDV values between the processed group and the control group.
Figures 3A et 3B : Figures 3A and 3B :
Graphe indiquant les valeurs de LVEF à 1 mois (3A) et LVEF à 2 mois (3B) selon les catégories de risque.Graph showing LVEF values at 1 month (3A) and LVEF at 2 months (3B) according to the risk categories.
Figure 4 : Figure 4 :
Régression linéaire présentant la corrélation entre l'amélioration fonctionnelle et le nombre de cellules injectées.Linear regression showing the correlation between functional improvement and the number of cells injected.
Figure 5 : Figure 5 :
Echocardiographies pré-opératoire (A) et post- opératoire (B) : On voit nettement l'épaississement systolique de la paroi postérieure initialement akimétique.Pre-operative (A) and post-operative (B) echocardiograms: The systolic thickening of the posterior ankemic wall is clearly visible.
Figure 6 : Figure 6 :
Vue horizontale des deux ventricules par imagerie par tomographie à émission de positron présentant la paroi postérieure du ventricule gauche (zone greffée en bas du scan). Les photos 6A et 6B (haut) montrent une activité métabolique homogène dans les parois septales et antérieures avec une diminution de l'activité métabolique dans la paroi postérieure avant l'opération. Les photos 6C et CD (bas) montrent l'assimilation du désoxyglucose 2fluoro18 dans la paroi postérieure après opération.Horizontal view of the two ventricles by positron emission tomography imaging showing the posterior wall of the left ventricle (grafted area at the bottom of the scan). Photos 6A and 6B (top) show a homogeneous metabolic activity in the septal and anterior walls with a decrease in metabolic activity in the posterior wall before the operation. Photos 6C and CD (bottom) show the uptake of deoxyglucose 2 fluoro 18 in the posterior wall after surgery.
Figure 7 : Figure 7 :
Stabilité des caractéristiques des populations cellulaires (CD56+) en fonction des expansions successivesStability of cell population characteristics (CD56 +) as a function of successive expansions
A. PROCEDE D'OBTENTION DE POPULATIONS CELLULAIRES ISSUES DE TISSUS MUSCULAIRES SQUELETTIQUESA. METHOD FOR OBTAINING CELLULAR POPULATIONS FROM SKELETAL MUSCLE TISSUES A.1 Matériels, solutions et milieux utilisés A.1 Materials, solutions and media used

Milieu A : Milieu MCDB120 (23) modifié : substitution de la L-Valine par de la D-Valine, élimination du rouge de phénol, élimination de la thymidine. Medium A: Modified MCDB120 medium (23): substitution of L-Valine with D-Valine, removal of phenol red, removal of thymidine.

Milieu B : Milieu A + 20% de sérum de veau foetal irradié + antibiotiques (à 100 Ul/ ml pour la pénicilline et 100 µg/ml pour la streptomycine). Medium B: Medium A + 20% irradiated fetal calf serum + antibiotics (at 100 IU / ml for penicillin and 100 μg / ml for streptomycin).

Milieu C : Milieu B + bFGF (10 ng/ml) + dexamethasone (1 µM). Medium C: Medium B + bFGF (10 ng / ml) + dexamethasone (1 μM).

Solution D : PBS Solution D: PBS

Milieu E : Milieu A + bFGF (10 ng/ml) + dexamethasone (1 µM) + sérumalbumine humaine stérile (0.5%). Medium E: Medium A + bFGF (10 ng / ml) + dexamethasone (1 μM) + sterile human serum albumin (0.5%).

Solution F : Solution saline isotonique stérile 0.9% NaCl. Solution F: Sterile isotonic saline solution 0.9% NaCl.

Solution G : Solution F + 4% de sérumalbumine humaine et 7.5% de DMSO (diméthyl-sulfoxyde) final. Solution G: Solution F + 4% human serum albumin and 7.5% final DMSO (dimethyl sulfoxide).

Solution H : Solution d'injection F + 0,5% de sérumalbumine humaine Solution H : Injection solution F + 0.5% human serum albumin

Le procédé d'obtention de populations cellulaires décrit ci-après comporte 6 étapes :

  • ■ prélèvement et éminçage de la biopsie
  • ■ dissociation enzymatique et séparation des cellules indivualisées par filtration
  • ■ mise en culture des cellules et phases d'expansion des cultures
  • ■ identification des types cellulaires présents aux différents stades de la culture par l'analyse de marqueurs cellulaires spécifiques
  • ■ récolte des cellules
  • ■ préparation et/ou maintien en survie et/ou congélation des cellules
  • ■ préparation du produit de thérapie cellulaire
The method for obtaining cell populations described below comprises 6 steps:
  • ■ sampling and cutting of the biopsy
  • ■ enzymatic dissociation and separation of indivualized cells by filtration
  • ■ cell culture and crop expansion phases
  • ■ identification of cell types present at different stages of the culture by the analysis of specific cell markers
  • ■ harvesting cells
  • ■ preparation and / or maintenance of the cells' survival and / or freezing
  • ■ preparation of the cell therapy product

Dans le procédé, la centrifugation des cellules s'effectue à 160 g pendant 5 minutes. Le comptage des cellules et l'analyse des populations sont réalisés à l'aide d'un hémacytomètre de Neubauer. Dans les phases d'expansion, les cellules sont incubées à 37°C dans un incubateur air-CO2 (95%-5%) saturé en humidité. L'observation des cellules se fait à l'aide d'un microscope inversé à contraste de phase.In the process, the cells are centrifuged at 160 g for 5 minutes. Cell counting and population analysis are performed using a Neubauer hemacytometer. In the expansion phases, the cells are incubated at 37 ° C. in an air-CO 2 incubator (95% -5%) saturated with moisture. The cells are observed using an inverted phase-contrast microscope.

A.2 Résultats A.2 Results A.2.1 Prélèvement et éminçage de la biopsieA.2.1 Biopsy sampling and slicing

Le prélèvement est réalisé au bloc opératoire, en milieu stérile, en système ouvert. Une biopsie d'environ 10-16 grammes de tissu musculaire squelettique est réalisée par l'équipe chirurgicale. La biopsie est ensuite découpée en petits cubes de 2 à 4 mm de côté, puis émincée à l'aide de ciseaux fins dans le milieu A. L'éminçat est placé dans une bouteille stérile contenant 25 ml de milieu A.The sample is taken in the operating room, in a sterile environment, in an open system. A biopsy of approximately 10-16 grams of skeletal muscle tissue is performed by the surgical team. The biopsy is then cut into small cubes of 2 to 4 mm on one side, then minced with fine scissors in medium A. The emine is placed in a sterile bottle containing 25 ml of medium A.

L'étape d'éminçage est aussi réalisé de manière assistée, à l'aide de broyeurs à couteaux Medimachine ® (distribué par Becton-Dickinson). Dans ce dispositif, des fragments de masse inférieure à 0,2g sont dissociés dans les réceptacles Medicon stériles, à l'issue de broyage contrôlé par un moteur électrique, d'une durée inférieure à 5 minutes. La répétition de l'opération à l'aide de plusieurs réceptacles Medicon permet de préparer finalement plusieurs grammes de muscle. Le tableau A ci-après présente les proportions des types cellulaires CD56+, CD15+ et CD34+ obtenues au cours des différents stades de culture J0, J15, J20 et J26 (la proportion de cellules CD34 étant quasi-nulle à J15, celle-ci n'est pas indiquée dans le tableau pour les étapes suivant la mise en culture J0). On observe que, de manière inattendue, l'étape d'éminçage de la biopsie constitue une étape cruciale pour obtenir rapidement une population comprenant un type cellulaire dominant de type myoblaste CD56+. TABLEAU A : COMPARAISON EMINCAGE AUX CISEAUX / BROYAT MECANIQUE BIOPSIE 1 BIOPSIE 2 EMINÇAGE AUX CISEAUX BROYAT MECANIQUE EMINÇAGE AUX CISEAUX BROYAT MECANIQUE J0 POIDS 0,8 g 0,8 g 1,2 g 1,4 g CNT 10*5 2,6 2,4 6,4 4,8 %CD34+ 7,4 4,7 2,6 1 %CD56+ 0,4 0 1,2 0,4 %CD15+ 0 0 0,9 3,3 PASSAGE 1 JOUR J15 J15 J7 J15 CNT 10*5 11,7 18,4 16,3 16,3 %CD56+ 51,6 90,5 35,1 77,9 %CD15+ 1,8 0,9 44,1 2,6 PASSAGE 2 JOUR J20 J20 J21 J21 %CD56+ 58 93,4 15 89,6 %CD15+ 20,9 4,4 20 7,3 PASSAGE 3 JOUR J26 J26 J27 J27 %CD56+ 43,6 75,8 49,7 60,2 %CD15+ 30,1 1,7 10,9 1,1 The slicing stage is also assisted, using Medimachine ® knife mills (distributed by Becton-Dickinson). In this device, fragments of mass less than 0.2 g are dissociated in the sterile Medicon receptacles, after grinding controlled by an electric motor, lasting less than 5 minutes. Repetition of the operation using several receptacles Medicon allows to finally prepare several grams of muscle. Table A below shows the proportions of the CD56 +, CD15 + and CD34 + cell types obtained during the different culture stages J0, J15, J20 and J26 (the proportion of CD34 cells being almost zero at J15, this one does not is not indicated in the table for the steps following the cultivation J0). It is observed that, unexpectedly, the biopsy cutting step is a crucial step in rapidly obtaining a population comprising a dominant CD56 + myoblast type cell type. TABLE A: SCISSING COMPILATION WITH SCISSORS / MECHANICAL BROYAT BIOPSY 1 BIOPSY 2 SCISSING WITH SCISSORS MECHANICAL BROYAT SCISSING WITH SCISSORS MECHANICAL BROYAT D0 WEIGHT 0.8 g 0.8 g 1.2 g 1.4 g CNT 10 * 5 2.6 2.4 6.4 4.8 % CD34 + 7.4 4.7 2.6 1 % CD56 + 0.4 0 1.2 0.4 % CD15 + 0 0 0.9 3.3 PASSAGE 1 DAY J15 J15 J7 J15 CNT 10 * 5 11.7 18.4 16.3 16.3 % CD56 + 51.6 90.5 35.1 77.9 % CD15 + 1.8 0.9 44.1 2.6 PASSAGE 2 DAY J20 J20 J21 J21 % CD56 + 58 93.4 15 89.6 % CD15 + 20.9 4.4 20 7.3 PASSAGE 3 DAY J26 J26 J27 J27 % CD56 + 43.6 75.8 49.7 60.2 % CD15 + 30.1 1.7 10.9 1.1

Différentes tailles de prélèvement ont été testées pour la mise en oeuvre du procédé de l'invention, couvrant une gamme de poids de 0,13 g à 14,9 g. Les résultats présentés dans les tableaux suivants montrent que l'évolution des proportions des différents types de population et l'amplification du nombre de cellules évoluent de manière comparable pour des tailles de biopsies de moins de 1 g (tableau B) à plus de 10 g (tableau C).

Figure imgb0001
Tableau C : Cultures à partir de biopsies de poids inférieur à 0,5 g PASSAGE 1 PASSAGE 2 PASSAGE 3 PASSAGE 1 PASSAGE 2 PASSAGE 3 BIOPSIE J TRAITEMENT J0 J10 J14 J16 BIOPSIE J TRAITEMENT J0 J6 J11 J17 5007 CNT 10*5 1,2 11,2 64 266 5054 CNT 10*5 2,8 6,6 7,1 370 % CD34+ 28,8 ND ND 0 % CD34+ 10,7 ND ND ND 0,23 g %CD56+ 1,95 68,6 87,6 89,5 0,45 g %CD56+ 25,7 62,3 69,1 84,6 %CD15+ ND 30,9 17,3 28,8 %CD15+ ND 31 26,9 14,5 % VIABILITE 88,9 96 100 98,7 % VIABILITE 88,7 100 96 87 BIOPSIE J TRAITEMENT J0 J7 J10 J15 BIOPSIE J TRAITEMENT J0 J7 J8 J15 5008 CNT 10*5 2,04 7 21,6 586,6 5058 CNT 10*5 7,7 9,4 13,6 534 % CD34+ 34 ND ND 0 % CD34+ 44,3 21,4 ND ND 0,23 g %CD56+ 6,1 47,1 66,3 92,9 0,19 g %CD56+ 8,4 23,2 27,5 72,2 %CD15+ ND 44,6 30,1 9,5 %CD15+ ND 63,2 63,6 18,9 % VIABILITE 85,1 92 88 97,9 % VIABILITE 95 82,1 100 97 BIOPSIE J TRAITEMENT J0 J7 J10 J14 BIOPSIE J TRAITEMENT J0 J6 J13 J15 5011 CNT 10*5 2,1 9,8 35 285 5060 CNT 10*5 5,2 9 18,8 663 % CD34+ 26,2 ND ND ND % CD34+ 48,7 ND ND ND 0,2 g %CD56+ 3,5 24,1 29,9 38 0,33 g %CD56+ 7,9 42,3 52,8 89,7 %CD15+ ND 70,8 63,8 60,5 %CD15+ ND 46,8 48,6 11,9 % VIABILITE 85,5 98 98 99 % VIABILITE 94,4 100 99 98 Different sample sizes were tested for carrying out the process of the invention, covering a weight range of 0.13 g to 14.9 g. The results presented in the following tables show that the evolution of the proportions of the different types of population and the amplification of the number of cells evolve in a comparable manner for biopsy sizes of less than 1 g (Table B) to more than 10 g (Table C).
Figure imgb0001
<b> Table C: Crops from biopsies weighing less than 0.5 g </ b> PASSAGE 1 PASSAGE 2 PASSAGE 3 PASSAGE 1 PASSAGE 2 PASSAGE 3 BIOPSY J TREATMENT D0 J10 J14 J16 BIOPSY J TREATMENT D0 J6 J11 J17 5007 CNT 10 * 5 1.2 11.2 64 266 5054 CNT 10 * 5 2.8 6.6 7.1 370 % CD34 + 28.8 ND ND 0 % CD34 + 10.7 ND ND ND 0.23 g % CD56 + 1.95 68.6 87.6 89.5 0.45 g % CD56 + 25.7 62.3 69.1 84.6 % CD15 + ND 30.9 17.3 28.8 % CD15 + ND 31 26.9 14.5 % VIABILITY 88.9 96 100 98.7 % VIABILITY 88.7 100 96 87 BIOPSY J TREATMENT D0 J7 J10 J15 BIOPSY J TREATMENT D0 J7 J8 J15 5008 CNT 10 * 5 2.04 7 21.6 586.6 5058 CNT 10 * 5 7.7 9.4 13.6 534 % CD34 + 34 ND ND 0 % CD34 + 44.3 21.4 ND ND 0.23 g % CD56 + 6.1 47.1 66.3 92.9 0.19 g % CD56 + 8.4 23.2 27.5 72.2 % CD15 + ND 44.6 30.1 9.5 % CD15 + ND 63.2 63.6 18.9 % VIABILITY 85.1 92 88 97.9 % VIABILITY 95 82.1 100 97 BIOPSY J TREATMENT D0 J7 J10 J14 BIOPSY J TREATMENT D0 J6 J13 J15 5011 CNT 10 * 5 2.1 9.8 35 285 5060 CNT 10 * 5 5.2 9 18.8 663 % CD34 + 26.2 ND ND ND % CD34 + 48.7 ND ND ND 0.2 g % CD56 + 3.5 24.1 29.9 38 0.33 g % CD56 + 7.9 42.3 52.8 89.7 % CD15 + ND 70.8 63.8 60.5 % CD15 + ND 46.8 48.6 11.9 % VIABILITY 85.5 98 98 99 % VIABILITY 94.4 100 99 98

Des biopsies ont été prélevées de patients âgés de 15 à 73 ans. Les résultats présentés dans le tableau D suivant montrent que le procédé est applicable quel que soit l'âge du patient sur lequel la biopsie est prélevée. Tableau D : Préparation de cellules d'origine musculaire à partir de biopsies provenant de patients de différents âges Jour 0 Premier passage Second passage Troisième passage Nom Poids (g) Age patient (années) Nombre 10*5 % CD56+ Nombre 10*6 % CD56+ Nombre 10*6 % CD56+ Nombre 10*6 % CD56+ MYO1 14.9 73 100 3.2 19.5 48 315 58.3 890 67.3 MYO3 10.4 63 43 3.4 14.2 67.7 156 87.1 922 91.3 MYO4 13.9 67 102 32.3 3.4 76.9 115 97.5 657 97.1 MYO5 11.6 39 117 22.1 31 71.5 244 89.9 993 95.2 MYO6 12 55 164 28.7 26.5 82 483 91.3 1210 84.9 4929 0.19 15 1 ND 1.6 64.3 4.7 75.4 56 82.4 5008 0.24 45 2 6.1 0.7 47.1 2.2 66.3 59 92.9 MOS 3.6 84 110 ND 75 93.4 240 96.4 565 93.7 CEL 3.0 51 45 ND 42 51.8 120 63.7 548 68.7 Biopsies were taken from patients aged 15 to 73 years. The results presented in Table D below show that the method is applicable regardless of the age of the patient on which the biopsy is taken. Table D: Preparation of muscle-derived cells from biopsies from patients of different ages Day 0 First passage Second passage Third passage Last name Weight (g) Patient age (years) Number 10 * 5 % CD56 + Number 10 * 6 % CD56 + Number 10 * 6 % CD56 + Number 10 * 6 % CD56 + MYO1 14.9 73 100 3.2 19.5 48 315 58.3 890 67.3 MYO3 10.4 63 43 3.4 14.2 67.7 156 87.1 922 91.3 MYO4 13.9 67 102 32.3 3.4 76.9 115 97.5 657 97.1 MYO5 11.6 39 117 22.1 31 71.5 244 89.9 993 95.2 MYO6 12 55 164 28.7 26.5 82 483 91.3 1210 84.9 4929 0.19 15 1 ND 1.6 64.3 4.7 75.4 56 82.4 5008 0.24 45 2 6.1 0.7 47.1 2.2 66.3 59 92.9 MOS 3.6 84 110 ND 75 93.4 240 96.4 565 93.7 CEL 3.0 51 45 ND 42 51.8 120 63.7 548 68.7

Les biopsies peuvent être conservées 90 h à 4°C ou congelées dans une solution saline équilibrée avant leur mise en culture. Les tableaux E et F suivants montrent que la viabilité des cellules et l'évolution des proportions des différents types de population né sont pas significativement affectées après 90 h de conservation de la biopsie à 4°C, ou après congélation Tableau E : Mise en culture d'une biopsie conservée 90 h à +4°C dans une solution saline équilibrée et préparation des cellules musculaires selon le procédé POIDS : 1,05 g J0 PASSAGE 1 (J7) CNT 10*6 0,8 4,9 % CD34+ 7,7 NS %CD56+ 6,5 58,7 %CD15+ 8,4 37,7 % VIABILITE 90,5 95 NS : non significatif Tableau F : Culture à partir d'une biopsie décongelée PASSAGE 1 PASSAGE 2 PASSAGE 3 BIOPSIE J TRAITEMENT J0 J18 J20 J25 4929 CNT 10*5 0,97 16,2 47,3 55?7 décongelée % CD34+ 20,5 ND 0,04 0 muscle non % CD56+ 44,9 64,3 75,4 82,4 pathologique % CD15+ ND 27,1 24,5 24,5 POIDS : 0,19 g % VIABILITE 90,8 100 96,8 96,7 Biopsies can be stored for 90 hours at 4 ° C or frozen in balanced saline before being cultured. The following Tables E and F show that the viability of the cells and the evolution of the proportions of the different types of population born are not significantly affected after 90 hours of preservation of the biopsy at 4 ° C., or after freezing. Table E: Placement of a preserved biopsy 90 h at + 4 ° C in a balanced saline solution and preparation of muscle cells according to the method WEIGHT: 1.05 g D0 PASSAGE 1 (J7) CNT 10 * 6 0.8 4.9 % CD34 + 7.7 NS % CD56 + 6.5 58.7 % CD15 + 8.4 37.7 % VIABILITY 90.5 95 NS: not significant PASSAGE 1 PASSAGE 2 PASSAGE 3 BIOPSY J TREATMENT D0 J18 J20 J25 4929 CNT 10 * 5 0.97 16.2 47.3 55? 7 thawed % CD34 + 20.5 ND 0.04 0 muscle not % CD56 + 44.9 64.3 75.4 82.4 pathological % CD15 + ND 27.1 24.5 24.5 WEIGHT: 0.19 g % VIABILITY 90.8 100 96.8 96.7

Le procédé de culture peut être mis en oeuvre à partir de biopsies de sujets sains ou de patients atteints de pathologie. Les résultats suivants présentés dans le tableau G montrent en particulier que le procédé de l'invention peut être mis en oeuvre pour la préparation de cellules d'origine musculaire issues d'un patient atteint de la dystrophie musculaire de Duchenne. Tableau G : Culture à partir d'une biopsie décongelée provenant d'un patient atteint de la dystrophie musculaire de Duchenne PASSAGE 1 PASSAGE 2 PASSAGE 3 PASSAGE 4 PASSAGE 5 BIOPSIE STADE J0 J7 J14 J21 J26 J29 4964 CNT 10*5 1,58 1,78 11,6 473,6 2411,2 6397 DE 24 H % CD34+ 53,9 ND 0,3 0 ND 0 muscle strié % CD56+ 5 24,6 78,1 73,7 60 52,5 paravertébral % CD15+ 0,4 30,4 12,7 15,5 26,4 36,1 POIDS : 0,136 g % VIABILITE ND 94 82,5 91,2 95,6 98 The culture method can be implemented from biopsies of healthy subjects or patients with pathology. The following results presented in Table G show in particular that the the invention can be implemented for the preparation of cells of muscular origin derived from a patient suffering from Duchenne muscular dystrophy. Table G: Culture from a thawed biopsy from a patient with Duchenne muscular dystrophy PASSAGE 1 PASSAGE 2 PASSAGE 3 PASSAGE 4 PASSAGE 5 BIOPSY STADIUM D0 J7 J14 J21 J26 J29 4964 CNT 10 * 5 1.58 1.78 11.6 473.6 2411.2 6397 DE 24 H % CD34 + 53.9 ND 0.3 0 ND 0 striated muscle % CD56 + 5 24.6 78.1 73.7 60 52.5 paravertébral % CD15 + 0.4 30.4 12.7 15.5 26.4 36.1 WEIGHT: 0.136 g % VIABILITY ND 94 82.5 91.2 95.6 98

Les résultats ci-après présentés montrent que le procédé peut être mis en oeuvre à partir de tout type de biopsies musculaires. En particulier, des biopsies ont été obtenues de différents muscles, le paravertébral, le jambier antérieur, le long péronier, l'extenseur commun orteils, le long péronier latéral, le jambier postérieur et le soléaire. Les résultats obtenus en fonction de différentes biopsies sont présentés dans le tableau H.

Figure imgb0002
The results presented below show that the method can be implemented from any type of muscle biopsies. In particular, biopsies were obtained from different muscles, the paravertebral, the anterior tibialis, the peroneal longus, the common extensor toes, the peroneal longus, the posterior tibialis and the soleus. The results obtained according to different biopsies are presented in Table H.
Figure imgb0002

A.2.2 Dissociation enzymatique et séparation des cellules individualisées par filtrationA.2.2 Enzymatic dissociation and separation of individualized cells by filtration

Le flacon contenant l'éminçat est centrifugé à température ambiante. Le surnageant est éliminé par aspiration. Le poids de l'éminçat est obtenu par pesée sur une balance tarée à l'aide d'un flacon vide. L'éminçat est rincé avec 25 ml du milieu A. Après sédimentation des éminçats, le surnageant est éliminé par aspiration.The vial containing the emine is centrifuged at room temperature. The supernatant is removed by aspiration. The weight of the emerald is obtained by weighing on a weighed scale using an empty bottle. The emulsion is rinsed with 25 ml of medium A. After sedimentation of the emulsions, the supernatant is removed by suction.

Une solution de libérase (Roche-Boehringer) a été préparée selon les instructions du fabricant puis reconditionnée et congelée à la concentration de 10 mg/ml. La libérase est décongélée extemporanément puis ajoutée à l'éminçat à concentration de 0.1 mg/ml, sous un volume de 10 mL par gramme de tissu. La bouteille est placée à l'étuve à 37°C pour une durée de 60 minutes. La bouteille est agitée manuellement toutes les 5 à 10 minutes (diffusion de l'enzyme et dissociation mécanique douce). La suspension est ensuite centrifugée. Le surnageant est éliminé par aspiration à la pipette. Ce premier produit de digestion est ensuite incubé dans une solution de trypsine à 0.25%. On utilise 10 ml de solution enzymatique par gramme de tissu initial. La suspension est digérée durant 20 minutes à 37°C à l'étuve, avec agitation manuelle toutes les 5 minutes. Elle est ensuite aspirée et refoulée au travers d'une pipette de 25 ml. 10% de sérum de veau foetal irradié (Hyclone) est alors ajouté pour la neutralisation des activités enzymatiques.A liberase solution (Roche-Boehringer) was prepared according to the manufacturer's instructions and then reconditioned and frozen at a concentration of 10 mg / ml. The liberase is defrosted extemporaneously and then added to the emulsion at a concentration of 0.1 mg / ml, in a volume of 10 ml per gram of tissue. The bottle is placed in an oven at 37 ° C for a period of 60 minutes. The bottle is agitated manually every 5 to 10 minutes (diffusion of the enzyme and gentle mechanical dissociation). The suspension is then centrifuged. The supernatant is removed by pipetting. This first digestion product is then incubated in a 0.25% trypsin solution. 10 ml of enzymatic solution per gram of initial tissue is used. The suspension is digested for 20 minutes at 37 ° C. in an oven, with manual agitation every 5 minutes. It is then aspirated and discharged through a 25 ml pipette. 10% irradiated fetal calf serum (Hyclone) is then added for the neutralization of enzymatic activities.

Le produit de digestion est filtré sur tamis de 100 µm, puis de 40 µm, sous pression de la gravité, afin de séparer les cellules dissociées des tissus résiduels. Un tamis par gramme de tissu est utilisé (Cell strainer de Falcon). Le filtrat est centrifugé à 300 g durant 5 minutes. Après élimination du surnageant, les culots sont lavés à l'aide du milieu B, puis centrifugés. Le surnageant est éliminé par aspiration. Le culot est ensuite repris dans 10 ml de milieu C. 100 µl de volume est prélevé pour comptage. Une aliquote est réservée pour estimation de la viabilité par cytofluorimétrie (iodure de propidium).The digestion product is filtered through a 100 μm sieve and then 40 μm under gravity pressure in order to separate the dissociated cells from the residual tissues. One sieve per gram of tissue is used (Falcon Cell strainer). The filtrate is centrifuged at 300 g for 5 minutes. After removal of the supernatant, the pellets are washed with medium B and then centrifuged. The supernatant is removed by aspiration. The pellet is then taken up in 10 ml of medium C. 100 μl of volume is taken for counting. An aliquot is reserved for estimating viability by cytofluorimetry (propidium iodide).

A.2.3 Mise en culture des cellules et phases d'expansion des culturesA.2.3 Cell culture and crop expansion phases

Après déconditionnement, les cellules sont transvasées dans le dispositif de culture. Le dispositif de culture est un plateau de culture (Nunc Single Tray) d'une surface de 600 cm2. Le remplissage du plateau se fait par un orifice prévu à cet effet et obturé par un bouchon stérile à usage unique. Les cultures sont incubées à 37°C dans une enceinte saturée en humidité en atmosphère contrôlée air-CO2 (95%-5%).After deconditioning, the cells are transferred to the culture device. The culture device is a culture tray (Nunc Single Tray) with an area of 600 cm 2 . The filling of the tray is done through a hole provided for this purpose and closed by a sterile single-use stopper. The cultures are incubated at 37 ° C. in a chamber saturated with humidity in an air-CO 2 controlled atmosphere (95% -5%).

Le lendemain de la mise en culture, une première vidange est réalisée afin d'éliminer les cellules mortes et les débris musculaires. Une poche vide est connectée à l'un des deux orifices du dispositif de culture. Le milieu est éliminé par gravité et remplacé par 120 ml de milieu C qui sont ajoutés à la culture. Le milieu C est renouvelé au bout de 120 à 192 heures. L'expansion est décidée lorsque le degré de confluence des cellules atteint 20% à 50% ou lorsque les premiers myotubes apparaissent (environ 8 jours après la mise en culture).The day after the planting, a first emptying is carried out in order to eliminate the dead cells and the muscular debris. An empty bag is connected to one of the two orifices of the culture device. The medium is removed by gravity and replaced with 120 ml of medium C which are added to the culture. Medium C is renewed after 120 to 192 hours. The expansion is decided when the degree of confluence of the cells reaches 20% to 50% or when the first myotubes appear (approximately 8 days after the culturing).

Après vidange du milieu, les cellules sont lavées par agitation manuelle douce à l'aide de 50 ml de solution D. La solution D est vidangée puis 20 ml de solution de trypsine irradiée (0.25%) sont ajoutés. Le flacon est mis à incuber 5 minutes à 37°C. Les cellules sont récoltées dans une poche de 40 ml. L'action de la trypsine est neutralisée par l'addition de 10% de sérum de veau foetal. Le sérum est injecté à l'intérieur de la poche à l'aide d'une seringue. Les cellules sont centrifugées. Le surnageant est éliminé par transvasement dans une autre poche de vidange, reliée par un connecteur stérile. Le culot est remis en suspension dans 30 ml de milieu C pour lavage des cellules puis les cellules sont centrifugées. Le surnageant est éliminé par transvasement dans une autre poche de vidange, reliée par un connecteur stérile. Le culot cellulaire est resuspendu dans 20 ml de milieu C. Un aliquot est prélevé pour numération et analyse des populations. La viabilité est estimée à l'aide d'un cytofluorimètre. Les cellules sont ensuite transférées dans une poche contenant 500 ou 750 ml de milieu C, puis ensemencées dans deux ou trois unités double-plateaux (double-tray de Nunc) d'une surface totale de 1200 ou 1800 cm2. Les cellules sont mises à incuber. Une troisième expansion est décidée lorsque le degré de confluence des cellules atteint 60% à 70% ou lorsque les premiers myotubes apparaissent. Pour cette série d'expansions, les boîtes multi-étagées de 10 plateaux (multi-tray de Nunc) sont utilisées. Après vidange du milieu, les cellules sont lavées à l'aide de 100 ml de solution D. Le décollement de la couche cellulaire et la dissociation enzymatique des cellules sont réalisés après vidange de la solution de lavage par l'addition de 50 ml de trypsine irradiée (0.25%) dans chaque plateau. Les préparations sont mises à incuber 5 minutes à 37°C. Les cellules sont récoltées dans des poches stériles de 300 à 600 ml. L'action de la trypsine est neutralisée par l'addition de 10% en volume de sérum de veau foetal. Les cellules sont centrifugées et le surnageant est éliminé par connexion à une poche de vidange.After emptying the medium, the cells are washed by gentle manual stirring with 50 ml of solution D. The solution D is drained and then 20 ml of irradiated trypsin solution (0.25%) are added. The flask is incubated for 5 minutes at 37 ° C. The cells are harvested in a 40 ml bag. The action of trypsin is neutralized by the addition of 10% fetal calf serum. The serum is injected inside the bag using a syringe. The cells are centrifuged. The supernatant is removed by transfer to another emptying bag, connected by a sterile connector. The pellet is resuspended in 30 ml of medium C for washing the cells and then the cells are centrifuged. The supernatant is removed by transfer to another emptying bag, connected by a sterile connector. The cell pellet is resuspended in 20 ml of medium C. An aliquot is taken for counting and analysis of the populations. Viability is estimated using a cytofluorimeter. The cells are then transferred to a bag containing 500 or 750 ml of medium C, then seeded in two or three double-tray units (Nunc double-tray) with a total surface of 1200 or 1800 cm 2 . The cells are incubated. A third expansion is decided when the degree of confluence of the cells reaches 60% to 70% or when the first myotubes appear. For this series of expansions, multi-stage boxes of 10 trays (Nunc multi-tray) are used. After emptying the medium, the cells are washed with 100 ml of solution D. The detachment of the cell layer and the enzymatic dissociation of the cells are carried out after draining the washing solution by the addition of 50 ml of trypsin irradiated (0.25%) in each tray. The preparations are incubated for 5 minutes at 37 ° C. The cells are harvested in sterile pockets of 300 to 600 ml. The action of trypsin is neutralized by the addition of 10% by volume of fetal calf serum. The cells are centrifuged and the supernatant is removed by connection to a drain bag.

Le culot cellulaire est resuspendu dans 50 ml de milieu C puis transféré dans un ou deux flacons contenant 1200 ou 2400 ml de milieu C par un connecteur stérile. Une ou deux boîtes de culture de 10 plateaux multi-étagés (multi-tray de Nunc) est ensemencée avec 1200 à 2400 ml de la préparation cellulaire. Le contrôle visuel de la croissance en plateaux multi-étagés est impossible. Par conséquent, une boite de culture Nunc stérile simple plateau (single-tray) est ensemencée aussi avec 110 ml de la préparation cellulaire. Ce dernier plateau permet un contrôle visuel quotidien de la culture et de l'état de confluence des cellules. Les cellules sont ensuite mises à incuber. La culture dure de 3 à 5 jours. La veille de la récolte des cellules, le milieu est éliminé par vidange dans une poche stérile et remplacé par un volume équivalent de milieu E. La récolte finale est décidée lorsque le degré de confluence des cellules atteint 90% ou lorsque les premiers myotubes apparaissent.The cell pellet is resuspended in 50 ml of medium C and then transferred into one or two vials containing 1200 or 2400 ml of medium C by a sterile connector. One or two culture dishes of 10 multi-stage trays (multi-tray Nunc) is seeded with 1200 to 2400 ml of the cell preparation. Visual control of growth in multi-stage trays is impossible. Therefore, a single sterile Nunc culture dish (single-tray) is also seeded with 110 ml of the cell preparation. This last plate allows a daily visual control of the culture and the state of confluence of the cells. Cells are then incubated. The culture lasts 3 to 5 days. The day before harvesting the cells, the medium is removed by emptying into a sterile bag and replaced by an equivalent volume of medium E. The final harvest is decided when the degree of confluence of the cells reaches 90% or when the first myotubes appear.

A.2.4 identification des types cellulaires présents aux différents stades de la culture par l'analyse de marqueurs cellulaires spécifiquesA.2.4 identification of cell types present at different stages of culture by analysis of specific cell markers

La caractérisation est basée sur l'analyse par cytofluorimétrie de flux (FACS). Des anticorps dirigés contre les antigènes humains de surface cellulaire suivants ont été utilisés : CD5, CD10, CD11, CD13, CD14, CD15, CD16, CD18, CD19, CD20, CD28, CD31, CD34, CD38, CD40, CD40-ligand, CD44, CD45, CD56, CD62, CD71, CD80, CD86, CD90, CD105, CD117, CD138. Des anticorps dirigés contre les structures antigéniques suivantes ont aussi été utilisés: CD138, HLA-Classe I, HLA-DR, ELAM, ICAM, LECAM, Stro-1, S-endo-1, VCAM, VLA2, VLA3, VLA4, VLA5, VLA6.Characterization is based on flow cytofluorometric analysis (FACS). Antibodies to the following human cell surface antigens were used: CD5, CD10, CD11, CD13, CD14, CD15, CD16, CD18, CD19, CD20, CD28, CD31, CD34, CD38, CD40, CD40-ligand, CD44 , CD45, CD56, CD62, CD71, CD80, CD86, CD90, CD105, CD117, CD138. Antibodies directed against the following antigenic structures have also been used: CD138, HLA-Class I, HLA-DR, ELAM, ICAM, LECAM, Stro-1, S-endo-1, VCAM, VLA2, VLA3, VLA4, VLA5, VLA6.

L'analyse de l'expression de la desmine, protéine intracellulaire est réalisée de la manière suivante :

  • Après mise en suspension dans du PBS, les cellules sont fixées et perméabilisées par l'addition de 10 volumes de méthanol à 4°C pendant 5 minutes, puis centrifugées. Après lavage du méthanol résiduel et centrifugation, les cellules sont reprises dans du PBS contenant l'anticorps dirigé contre la desmine (Dako, clone D33, 1/100) et incubées durant 15 minutes. Après lavage et centrifugation, les cellules sont incubées durant 15 minutes en présence de l'anticorps secondaire dirigé contre l'anticorps primaire, couplé à un fluorophore. Après lavage et centrifugation, les cellules sont analysées par FACS. Le tableau I ci-après présente les caractéristiques des marqueurs principaux utilisés et les types cellulaires correspondants (types cellulaires).
The analysis of the expression of desmin, intracellular protein is carried out as follows:
  • After suspending in PBS, the cells are fixed and permeabilized by the addition of 10 volumes of methanol at 4 ° C for 5 minutes and then centrifuged. After washing the residual methanol and centrifugation, the cells are taken up in PBS containing the antibody directed against desmin (Dako, clone D33, 1/100) and incubated for 15 minutes. After washing and centrifugation, the cells are incubated for 15 minutes in the presence of the secondary antibody directed against the primary antibody, coupled to a fluorophore. After washing and centrifugation, the cells are analyzed by FACS. Table I below presents the characteristics of the main markers used and the corresponding cell types (cell types).

CARACTERISTIQUES CLASSIQUES DES MARQUEURS UTILISESCLASSICAL CHARACTERISTICS OF MARKERS USED

Tableau I : Présentation de certains marqueurs cellulaires utilisés dans le procédé et les types cellulaires les exprimant. Table I : Presentation of certain cellular markers used in the process and the cell types expressing them. MARQUEURMARKER PEN TYPES CELLULAIRESCELLULAR TYPES CD10CD10 lymphocytes préB. neutrophilespreB lymphocytes. neutrophil CD13CD13 monocytes, cellules myéloïdesmonocytes, myeloid cells CD15CD15 monocytes, macrophages, granulocytes, éosinophilesmonocytes, macrophages, granulocytes, eosinophils CD16CD16 NK, sous pop. lymphocytes T, neutrophilesNK, under pop. T lymphocytes, neutrophils CD34CD34 progéniteursprogenitors CD38CD38 LT activités, cellule souche, sous pop. L T,B,NKLT activities, stem cell, under pop. L T, B, NK CD40CD40 lymphocytes T CD4+ activésactivated CD4 + T cells CD44CD44 Anti HCAMAnti HCAM CD45CD45 leucocytesleukocytes CD56CD56 NK, sous pop. Lymphocytes TNK, under pop. T cells CD71CD71 cellules proliférantesproliferating cells CD117CD117 progéniteursprogenitors HLA CL1HLA CL1 antigène MHC classe 1MHC class 1 antigen HLA CL2HLA CL2 antigène MHC classe 2MHC class 2 antigen VLA3Vla3 lymphocytes BB lymphocytes VLA5VLA5 lymphocytes T mémoire, monocytes, plaquettesmemory T cells, monocytes, platelets VLA6VLA6 thymocytes, lymphocytes T mémoire, monocytesthymocytes, memory T cells, monocytes DESMINEdesmin cellules musculairesmuscle cells

L'analyse des populations cellulaires est réalisée aux différents stades de culture :

  • Au jour J0 correspondant à la mise en culture de la population fraîchement préparée.
The analysis of the cell populations is carried out at the different stages of culture:
  • Day D0 corresponding to the cultivation of the freshly prepared population.

Plusieurs séries ont été réalisées en flacons unitaires mis en culture parallèlement à l'unité simple plateau à J0, afin de pouvoir suivre quotidiennement l'évolution des cultures en prélevant un ou plusieurs flacons par jour.

  • Au jour 1 (J1), la fraction non adhérente (surnageant) et la fraction adhérente (obtenue par trypsination) ont été analysées indépendamment. De manière intéressante, il a été montré que ces deux fractions renferment des populations aux caractéristiques différentes.
  • De J2 à J9, les cellules adhérentes ont été analysées chaque jour afin de suivre l'évolution des types cellulaires au cours du temps.
Several series were carried out in unit bottles cultured parallel to the single plate unit at D0, in order to be able to monitor the evolution of the cultures daily by taking one or more bottles per day.
  • On day 1 (D1), the non-adherent fraction (supernatant) and the adherent fraction (obtained by trypsination) were analyzed independently. Interestingly, these two fractions have been shown to contain populations with different characteristics.
  • From day 2 to day 9, the adherent cells were analyzed daily to follow the evolution of cell types over time.

En parallèle, les cellules obtenues par production en masse ont été analysées à chaque étape d'expansion puis au moment de la récolte finale. Les résultats ont montré qu'à date de récolte équivalente, les caractéristiques obtenues dans les plateaux et les flacons indépendants sont identiques.In parallel, the cells obtained by mass production were analyzed at each expansion stage and then at the time of the final harvest. The results showed that at equivalent harvest date, the characteristics obtained in the trays and the independent flasks are identical.

Les données issues de l'identification des types cellulaires au cours de la cinétique de différenciation sont présentées dans les tableaux JA et JB suivants et les caractéristiques essentielles sont décrites ci-après.Data from the identification of cell types during differentiation kinetics are presented in the following Tables JA and JB and the essential characteristics are described below.

Cinétique de différenciation (pourcentages)Kinetics of differentiation (percentages)

Tableau JA : Identification des types cellulaires au cours du temps (en pourcentage dans la population cellulaire totale) des stades J0 à J3 de la culture. Table JA : Identification of cell types over time (as a percentage in the total cell population) of stages J0 to J3 of the culture. MARQUEURSMARKERS J0D0 J1J1 SN J1SN J1 J2J2 J3J3 CD34+CD34 + 38,5 (25-75)38.5 (25-75) 77 (69-79)77 (69-79) 39 (25-67)39 (25-67) 74 (64-75)74 (64-75) 74 (51-75)74 (51-75) CD34-CD34- 61,5 (25-75)61.5 (25-75) 23 (20-30)23 (20-30) 61 (33-75)61 (33-75) 26 (25-30)26 (25-30) 26 (25-49)26 (25-49) CD34+CD10+CD34 + CD10 + 14 (10-18)14 (10-18) 27 (25-29)27 (25-29) 2,5 (2-3)2.5 (2-3) 24 (20-40)24 (20-40) 11 (7-17)11 (7-17) CD34+CD10-CD34 + CD10- 25 (23-31)25 (23-31) 47 (40-54)47 (40-54) 30,5 (24-37)30.5 (24-37) 39 (36-54)39 (36-54) 59 (44-63)59 (44-63) CD34+CD45+CD34 + CD45 + <5<5 NDND 0,5 (0-1)0.5 (0-1) NDND NDND CD34+CD56+CD34 + CD56 + 00 00 00 0,3 (0,3-0,3)0.3 (0.3-0.3) 0,7 (0,4-2)0.7 (0.4-2) CD34+DR+CD34 + DR + 1414 NDND 21,5 (16-27)21.5 (16-27) NDND NDND CD34+DR-CD34 + DR- 2828 NDND 12 (12-12)12 (12-12) NDND NDND CD34+CD 15+CD34 + CD 15+ NDND NDND NDND NDND NDND CD56+CD56 + 4,7 (0-26)4.7 (0-26) 4,4 (0,3-15)4.4 (0.3-15) 1 (0-5)1 (0-5) 11 (7,7-20)11 (7.7-20) 14 (12-35)14 (12-35) CD15+CD15 + 1,5 (0-6)1.5 (0-6) 3 (1,5-6)3 (1,5-6) 3,6 (0,1-12)3.6 (0.1-12) 20,5 (20-21)20.5 (20-21) 25 (24-33)25 (24-33) CD56+CD15+CD56 + CD15 + 0,02 (0-0,3)0.02 (0-0.3) 00 00 4 (4-4)4 (4-4) 10 (6-14)10 (6-14) CD13+CD13 + NDND NDND NDND NDND NDND CD44+CD44 + 00 NDND NDND NDND NDND CD117+CD117 + 1,7 (0,16-1,7)1.7 (0.16-1.7) NDND 1,7 (0,2-2)1.7 (0.2-2) NDND NDND Tableau JB : Identification des types cellulaires au cours du temps (en pourcentage dans la population cellulaire totale) des stades J4 à J8 de la culture. Table JB : Identification of cell types over time (as a percentage in the total cell population) of the J4 to D8 stages of the culture. MARQUEURSMARKERS J4J4 J5J5 J6J6 J7J7 J8J8 CD34+CD34 + 56 (36-67)56 (36-67) 35 (26-50)35 (26-50) 23 (13-47)23 (13-47) 2,7 (0,6-10)2.7 (0.6-10) 1,7 (0,4-3,4)1.7 (0.4-3.4) CD34-CD34- 44 (33-63)44 (33-63) 65 (50-73)65 (50-73) 76 (53-87)76 (53-87) 97 (90-99)97 (90-99) 98 (96-100)98 (96-100) CD34+CD10+CD34 + CD10 + 9 (8-22)9 (8-22) 16,5 (3-34)16.5 (3-34) 16,8 (0,6-33)16.8 (0.6-33) 1,7 (0,2-2,7)1.7 (0.2-2.7) 0,4 (0,2-7,6)0.4 (0.2-7.6) CD34+CD10-CD34 + CD10- 35 (27-59)35 (27-59) 25 (15-35)25 (15-35) 14 (14-14)14 (14-14) 5,1 (0,4-9)5.1 (0.4-9) 1,3 (0,2-7,6)1.3 (0.2-7.6) CD34+CD45+CD34 + CD45 + NDND NDND NDND NDND NDND CD34+CD56+CD34 + CD56 + 3 (1,5-6)3 (1,5-6) 7,5 (1,5-12)7.5 (1.5-12) 3 (0,1-12)3 (0.1-12) 0,35 (0,2-0,6)0.35 (0.2-0.6) 0,4 (0,4-2,1)0.4 (0.4-2.1) CD34+DR+CD34 + DR + NDND NDND NDND NDND NDND CD34+DR-CD34 + DR- NDND NDND NDND NDND NDND CD34+CD15+CD34 + CD15 + NDND NDND NDND NDND NDND CD56+CD56 + 34 (28-55)34 (28-55) 61 (50-68)61 (50-68) 73 (57-74)73 (57-74) 64 (52-87)64 (52-87) 68,4 (50,3-91)68.4 (50.3-91) CD15+CD15 + 48 (46-48)48 (46-48) 48 (47-63)48 (47-63) 51 (29-64)51 (29-64) 36 (20-55)36 (20-55) 29 (12-54)29 (12-54) CD56+CD15+CD56 + CD15 + 19 (17-21)19 (17-21) 13 (9-25)13 (9-25) 10(5-13)10 (5-13) 3,6 (2,6-6)3.6 (2.6-6) 1,4 (1-3)1.4 (1-3) CD13+CD13 + NDND NDND 96 (76-97)96 (76-97) 99 (94-99)99 (94-99) 99 (96-99)99 (96-99) CD44+CD44 + NDND NDND NDND NDND NDND CD117+CD117 + NDND NDND NDND NDND NDND

A.2.4. 1 Caractéristiques des préparations cellulaires à J0 A.2.4. 1 Characteristics of cell preparations at day 0

Le procédé permet d'obtenir à J0 3x105 à 4X106 cellules par gramme de tissu. Les types cellulaires majoritaires sont CD34+. Les types cellulaires CD34+ sont CD34+/CD10+ à 14%; CD34+/CD10- à 25% ; CD34+/CD56+ à 0% ; CD34+/DR+ à 14% et CD34+/DR- à 28%. Les populations CD34+ sont CD45- en majorité. Des populations minoritaires expriment CD44, CD45, CD56, CD117, HLA-Classe Il. De manière surprenante, la caractérisation d'un nombre de cellules progénitrices important (en quantités absolue et relative dans la population) permet d'envisager, par une récolte des cellules à ce stade, leur utilisation comme produit de thérapie cellulaire pour la reconstitution de nombreux tissus, non seulement musculaires mais aussi hématopoïétiques, osseux, adipeux, cartilagineux ou vasculaires.The method makes it possible to obtain at 3 × 10 5 to 4 × 10 6 cells per gram of tissue. The majority cell types are CD34 +. The CD34 + cell types are CD34 + / CD10 + at 14%; CD34 + / CD10- at 25%; CD34 + / CD56 + at 0%; CD34 + / DR + at 14% and CD34 + / DR at 28%. The CD34 + populations are predominantly CD45-. Minority populations express CD44, CD45, CD56, CD117, HLA-Class II. Surprisingly, the characterization of a large number of progenitor cells (in absolute and relative amounts in the population) makes it possible to envisage, by harvesting the cells at this stage, their use as a cell therapy product for the reconstitution of numerous cells. tissues, not only muscle but also hematopoietic, bone, adipose, cartilaginous or vascular.

A.2.4.2 Caractéristiques des préparations cellulaires adhérentes à J1A.2.4.2 Characteristics of adherent cell preparations at J1

Une majorité de cellules est CD34+. La population se compose de CD34+/CD10+ (27%) et CD34+/CD10- (47%). CD13 apparaît. La préparation est négative pour CD117 et CD45. Les types cellulaires CD15+ et CD56+ sont minoritaires.A majority of cells is CD34 +. The population consists of CD34 + / CD10 + (27%) and CD34 + / CD10- (47%). CD13 appears. The preparation is negative for CD117 and CD45. The CD15 + and CD56 + cell types are in the minority.

A.2.4.3 Caractéristiques des préparations cellulaires présentes dans le surnageant à J1. A.2.4.3 Characteristics of cell preparations present in the supernatant on D1.

Une proportion importante de cellules est CD34+. La population se compose essentiellement de CD34+/CD10-. Une population CD117+ (<5%) est présente et exprime CD45+. Certaines populations minoritaires sont présentes: CD38+ (15%), CD45+, peu de CD15+ et de CD56+, HLA-Classe II+.A significant proportion of cells is CD34 +. The population consists mainly of CD34 + / CD10-. A CD117 + population (<5%) is present and expresses CD45 +. Some minority populations are present: CD38 + (15%), CD45 +, few CD15 + and CD56 +, HLA-Class II +.

A.2.4.4 Caractéristiques des évolutions des marqueurs au cours de la cultureA.2.4.4 Characteristics of changes in markers during culture

L'évolution est marquée par une augmentation croissante de la proportion de cellules CD15+ et/ou CD56+ et une diminution des populations CD34+. On peut observer au cours du temps la transition progressive d'une population de CD34+ à CD15+. La proportion des populations CD13+, CD44+, CD71+ augmente avec le temps. On observe des populations minoritaires CD138+, HLA-Classe II+, CD38+.The evolution is marked by an increasing increase in the proportion of CD15 + and / or CD56 + cells and a decrease in CD34 + populations. The gradual transition from a CD34 + to CD15 + population can be observed over time. The proportion of CD13 +, CD44 +, CD71 + populations increases with time. There are minority populations CD138 +, HLA-Class II +, CD38 +.

En fin d'expansion, on observe trois populations prépondérantes : CD56+, CD15+ et CD56-/CD15-. La population CD56+ exprime CD10, CD13, CD44, la desmine et HLA-Classe I. Ces marqueurs sont spécifiques des cellules myoblastiques. La population CD15+ exprime CD13, Classe I et partiellement CD10. Dans la population CD56-/CD15-, une fraction exprime la desmine et l'autre fraction ne l'exprime pas. Certains marqueurs sont exprimés plus faiblement et de façon variable : CD71, VLA3, VLA5, VLA6, CD16+ et CD40L. Les populations CD34+, CD38+, CD45+ et HLA-Classe II+ ont disparu ou sont extrêmement minoritaires.At the end of the expansion, there are three predominant populations: CD56 +, CD15 + and CD56- / CD15-. The CD56 + population expresses CD10, CD13, CD44, desmin and HLA-Class I. These markers are specific for myoblastic cells. The CD15 + population expresses CD13, Class I and partially CD10. In the CD56- / CD15- population, one fraction expresses desmin and the other fraction does not express it. Some markers are expressed more weakly and variably: CD71, VLA3, VLA5, VLA6, CD16 + and CD40L. The CD34 +, CD38 +, CD45 + and HLA-Class II + populations have disappeared or are extremely minor.

A.2.4.5 Caractéristiques des cellules après déplétion de la fraction CD34+ A.2.4.5 Cell characteristics after depletion of the CD34 + fraction

Le tableau K ci-dessous représente les caractéristiques des cellules obtenues, à l'issue du premier passage, en comparant différentes conditions initiales. Huit expériences indépendantes sont représentées. Après déplétion de la fraction CD34+, le procédé permet d'obtenir rapidement une population cellulaire très majoritairement composée de cellules exprimant le CD56. En particulier, la proportion de cellules exprimant CD56 est supérieure à celle obtenue à partir d'une biopsie non déplétée. Tableau K. Déplétions ou enrichissements en cellules CD34+ présentes au sein de la biopsie musculaire. %CD56+ au premier passage Expérience 1 Expérience 2 Expérience 3 Expérience 4 Expérience 5 Expérience 6 Expérience 7 Expérience 8 Fraction Non Séparée ND 72% 87 % 41 % 48 % 67 % 70% 38 % Fraction Déplétée en CD34 93 % 98 % 97 % 82% 93% 93 % 86% 57 % Fraction Enrichie en CD34 61 % 36% 37 % 1% 8% 28 % 4 % 12% Table K below represents the characteristics of the cells obtained at the end of the first pass, by comparing different initial conditions. Eight independent experiments are represented. After depletion of the CD34 + fraction, the method makes it possible to rapidly obtain a cell population that is predominantly composed of cells expressing CD56. In particular, the proportion of cells expressing CD56 is greater than that obtained from an unexplained biopsy. <u> Table K. Depletions or enrichments in CD34 + cells present in muscle biopsy. </ u> % CD56 + at first pass Experience 1 Experience 2 Experience 3 Experience 4 Experience 5 Experience 6 Experience 7 Experience 8 Non Separated Fraction ND 72% 87% 41% 48% 67% 70% 38% Fraction Deployed in CD34 93% 98% 97% 82% 93% 93% 86% 57% Fraction Enriched with CD34 61% 36% 37% 1% 8% 28% 4% 12%

A.2.5 Récolte des cellulesA.2.5 Harvesting cells

Le protocole suivant décrit la récolte des cellules au stade final pour l'obtention d'une population myoblastique majoritaire. Cependant, le protocole permet à l'homme du métier de le mettre en oeuvre quelque soit le stade de différenciation choisi pour la récolte des cellules et ce, en fonction de la population cellulaire recherchée.The following protocol describes harvesting the cells at the final stage to obtain a majority myoblastic population. However, the protocol allows the person skilled in the art to implement it whatever the stage of differentiation chosen for harvesting the cells and this, depending on the desired cell population.

Après vidange du milieu, les cellules sont lavées à l'aide de 500 ml de solution D (pour les unités multi-plateaux), 50 ml pour l'unité simple et 100 ml pour les unités doubles. La solution de lavage est vidangée et 200 ml de solution de trypsine irradiée (0.25%) sont ajoutés aux unités multi-plateaux (20 ml pour l'unité simple, 40 ml pour les unités doubles). La préparation est mise à incuber 5 minutes à 37°C. Les cellules sont récoltées par vidange dans une poche de 500 ml. L'action de la trypsine est neutralisée par l'addition de 10% de sérum de veau foetal injecté par une seringue. Les cellules sont lavées de la manière suivante : les cellules sont centrifugées. Le surnageant est éliminé puis les cellules sont remises en suspension dans 300 ml de solution F puis centrifugées. Les surnageants sont éliminés. Deux autres étapes de lavages comme précédemment décrit sont réalisées. Ces lavages successifs ont pour but d'éliminer la trypsine, les protéines animales encore présentes et le bFGF recombinant. Au cours du troisième lavage, une aliquote est réservée pour le comptage cellulaire, l'estimation de la viabilité et de la qualité cellulaire, les contrôles de qualité microbiologique. Les cellules peuvent être concentrées dans la solution H de manière à obtenir une suspension adéquate pour l'usage clinique demandé.After emptying the medium, the cells are washed with 500 ml of solution D (for the multi-tray units), 50 ml for the single unit and 100 ml for the double units. The wash solution is drained and 200 ml of irradiated trypsin solution (0.25%) is added to the multi-tray units (20 ml for the single unit, 40 ml for the double units). The preparation is incubated for 5 minutes at 37 ° C. The cells are collected by emptying in a 500 ml bag. The action of trypsin is neutralized by the addition of 10% fetal calf serum injected by a syringe. The cells are washed as follows: the cells are centrifuged. The supernatant is removed and the cells are resuspended in 300 ml of solution F and centrifuged. Supernatants are eliminated. Two other washing steps as previously described are carried out. These successive washes are intended to remove trypsin, animal proteins still present and recombinant bFGF. During the third wash, an aliquot is reserved for cell count, viability and cell quality estimation, microbiological quality controls. The cells can be concentrated in solution H so as to obtain a suspension that is adequate for the clinical use requested.

Après centrifugation, les cellules sont reprises dans un volume de solution isotonique à une concentration de 1.5x108 cellules/ml. Elles sont enfin aspirées dans une seringue de 10 ml. Les cellules sont prélevées pour l'injection dans des seringues stériles. Le type d'aiguille utilisée pour l'injection dépend du tissu ciblé. Pour l'injection intramyocardique directe, une aiguille coudée à 90° de 25 à 30 gauge est spécifiquement utilisée.After centrifugation, the cells are taken up in a volume of isotonic solution at a concentration of 1.5 × 10 8 cells / ml. They are finally sucked into a 10 ml syringe. The cells are removed for injection into sterile syringes. The type of needle used for the injection depends on the targeted tissue. For direct intramyocardial injection, a 90 ° 25 to 30 gauge bent needle is specifically used.

A.2.6 Rendements de production et caractéristiques des types cellulairesA.2.6 Production yields and characteristics of cell types

Le tableau L ci-dessous récapitule les résultats obtenus lors de la mise en oeuvre du procédé de l'invention à partir de différentes biopsies obtenues de trois patients différents.Table L below summarizes the results obtained during the implementation of the method of the invention from different biopsies obtained from three different patients.

Les cellules ont été produites initialement dans les unités de simples, doubles et multiples plateaux jusqu'à la troisième expansion incluse. Les expansions ont par la suite été réalisées par dédoublement des populations et repiquages dans des boites de culture de 25 cm2. A chaque passage, la majorité des cellules était utilisée pour la caractérisation et le comptage, tandis qu'un nombre connu de cellules était utilisé pour l'ensemencement de dédoublement. Le nombre de dédoublements cumulés permet, par le calcul, d'obtenir environ 100 milliards de cellules entre la huitième et la neuvième expansion.The cells were initially produced in single, double and multiple plate units until the third included expansion. The expansions were subsequently carried out by duplicating the populations and transplanting them into culture dishes of 25 cm 2 . At each pass, the majority of the cells were used for characterization and counting, while a known number of cells were used for duplicate seeding. The number of cumulative doublings allows, by calculation, to obtain about 100 billion cells between the eighth and the ninth expansion.

Le tableau L présente les rendements en terme de nombre de cellules obtenues et en terme de proportion de cellules du type CD56+ ou desmine+ dans la population aux différentes phases d'expansion pour les trois patients nommés MYO 003, MYO 004 et MYO 005.

Figure imgb0003
L'histogramme à la figure 7 présente les valeurs médianes d'expression de CD56 et de CD15 sur 8 échantillons au cours des différentes phases d'expansions.Table L shows the yields in terms of number of cells obtained and in terms of proportion of CD56 + or desmin + cells in the population at the different expansion phases for the three patients named MYO 003, MYO 004 and MYO 005.
Figure imgb0003
The histogram at the figure 7 presents the median expression values of CD56 and CD15 on 8 samples during the different phases of expansions.

Les résultats présentés à la figure 7 montrent que les proportions des types cellulaires CD56+ et CD15+ obtenues sont relativement homogènes selon les différentes biopsies, et ne sont pas modifiées au cours des différentes expansions. En particulier, on constate que les cellules de type CD56+ restent en proportion dominantes au cours des phases d'expansion.The results presented at figure 7 show that the proportions of the CD56 + and CD15 + cell types obtained are relatively homogeneous according to the different biopsies, and are not modified during the different expansions. In particular, it is found that the CD56 + type cells remain in a dominant proportion during the expansion phases.

Ces résultats montrent en outre, que après identification des stades de récoltes optimaux en fonction des types cellulaires recherchés, l'homme du métier peut reproduire le procédé de l'invention en s'affranchissant de l'étape de caractérisation cellulaire telle que définie dans le procédé de l'invention et en multipliant les phases d'expansion de manière à obtenir un grand nombre de cellules comprenant un type cellulaire spécifique en proportion dominante, et notamment le type cellulaire CD56+.These results further show that after identification of optimal harvest stages according to the desired cell types, a person skilled in the art can reproduce the process of the invention by dispensing with the cell characterization step as defined in FIG. method of the invention and by multiplying the expansion phases so as to obtain a large number of cells comprising a specific cell type in a dominant proportion, and in particular the CD56 + cell type.

A.2.7 Congélation du produit de thérapie cellulaireA.2.7 Freezing the cell therapy product

Afin de permettre l'utilisation dans le temps des cellules ainsi préparées, il peut être avantageux de les congeler dans des conditions telles que la décongélation ultérieure permette une survie des cellules suffisante, de préférence supérieure à 90%.In order to allow the cells thus prepared to be used over time, it may be advantageous to freeze them under conditions such that subsequent thawing allows sufficient cell survival, preferably greater than 90%.

A titre d'exemple, les cellules sont suspendues dans le milieu de congélation (solution G) et transférées dans deux poches de congélation stériles, à une concentration comprise entre 107 et 2x107 cellules/ml ou dans des tubes de cryocongélation à une concentration comprise entre 1 x 106 et 5 x 106/ml. La congélation est réalisée à l'aide d'un dispositif (Digicool ou Nicool) assurant une descente progressive en température contrôlée. Les cellules sont stockées dans l'azote liquide jusqu'au moment de la décongélation.By way of example, the cells are suspended in the freezing medium (solution G) and transferred to two sterile freezing bags at a concentration of between 10 7 and 2 × 10 7 cells / ml or in cryocongesting tubes at a concentration of between 1 x 10 6 and 5 x 10 6 / ml. The freezing is carried out using a device (Digicool or Nicool) ensuring a progressive descent in temperature controlled. The cells are stored in liquid nitrogen until defrosting.

La décongélation des cellules est réalisée au bain-marie à 37°C. Les préparations cellulaires sont lavées deux fois à l'aide d'une solution saline isotonique. Les rinçages sont effectués par connexion stérile aux poches de solution isotonique et aux poches de vidange. Une aliquote est réservée pour l'estimation de la viabilité et de la qualité cellulaire.Thawing of the cells is carried out in a water bath at 37 ° C. Cell preparations are washed twice with isotonic saline. The rinses are performed by sterile connection to the pockets of isotonic solution and the emptying pockets. An aliquot is reserved for estimating viability and cell quality.

B. FACTEURS AFFECTANT LA FONCTIONNALITE DE LA TRANSPLANTATION DE CELLULES D'ORIGINE MUSCULAIRE AUTOLOGUES POUR LE TRAITEMENT DE MODELES D'ISCHEMIES MYOCARDIQUESB. FACTORS AFFECTING THE FUNCTIONALITY OF TRANSPLANTATION OF AUTOLOGOUS MUSCLE CELLS FOR THE TREATMENT OF MYOCARDIAL ISCHEMIC MODELS B.1 Matériels et méthodes B.1 Materials and methods B.1.1 Modèle d'ischémie myocardiqueB.1.1 Model of myocardial ischemia

Des rats mâles Wistar, pesant 280 g ont été anesthésiés à la kétamine (50 mg/kg) et la xylasine (10 mg/kg) et ventilés par la trachée. Une thoracotomie a été réalisée. L'infarctus du myocarde est obtenu par ligature coronaire gauche en utilisant un fil de polypropylène 7/0.Wistar male rats weighing 280 g were anesthetized with ketamine (50 mg / kg) and xylasin (10 mg / kg) and ventilated by the trachea. A thoracotomy was performed. Myocardial infarction is obtained by left coronary ligation using 7/0 polypropylene wire.

B.1.2 Tests de fonctionnalitéB.1.2 Functionality tests

Une semaine après l'infarctus du myocarde, et un ou deux mois après transplantation, la fonction ventriculaire gauche a été étudiée par échocardiographie bi-dimensionnelle (2D).One week after myocardial infarction, and one or two months after transplantation, left ventricular function was studied by two-dimensional (2D) echocardiography.

Sous anesthésie générale à la kétamine (50 mg/kg) et la xylasine (10 mg/kg), les mesures 2D (et M-mode) sont réalisées avec un appareil commercial de 15MHz (15L8) « linear-array transducer » (Sequoia, Acuson Corp., Mountain View, CA, USA) autorisant une fréquence maximale de 160 Hz. Des vues parasternales longitudinales sont obtenues, de manière à ce que les valves mitrales et aortiques et l'apex puissent être correctement visualisées et ainsi enregistrées.Under general anesthesia with ketamine (50 mg / kg) and xylasin (10 mg / kg), the 2D (and M-mode) measurements are made with a 15MHz (15L8) linear-array transducer (Sequoia) , Acuson Corp., Mountain View, CA, USA) allowing a maximum frequency of 160 Hz. Longitudinal parasternal views are obtained, so that the mitral and aortic valves and the apex can be correctly visualized and thus recorded.

Les mesures des longueurs (L) du grand axe du ventricule gauche et les tracés des zones endocardiaques (a) sont réalisées. Le volume en fin de diastole du ventricule gauche (LVEDV) et le volume en fin de systole du ventricule gauche (LVESV) ont été calculés avec la formule suivante : V = 8x A2 / (3 x π x L). Les fractions d'éjection du ventricule gauche (LVEF) sont ainsi calculées : LVEF=(LVEDV-LVESV)/LVEDV. Toutes les mesures ont été faites à partir d'au moins trois battements et à l'aide de deux expérimentateurs traitant les différents groupes en aveugle.The length measurements (L) of the long axis of the left ventricle and the plots of the endocardial zones (a) are carried out. The end diastolic volume of the left ventricle (LVEDV) and the volume at the end of left ventricular systole (LVESV) were calculated with the following formula: V = 8x A 2 / (3 x π x L). The left ventricular ejection fractions (LVEF) are thus calculated: LVEF = (LVEDV-LVESV) / LVEDV. All measurements were made from at least three beats and with the aid of two experimenters treating the different groups blindly.

B.1.3 Culture des cellulesB.1.3 Cell culture

Pendant la procédure d'infarctus du myocarde, les muscles antérieurs des tibiaux droit et gauche sont disséqués de manière à séparer le tendon et le tissu aponeurotique du tissu musculaire. Ils sont ensuite émincés, pesés et dissociés à l'aide d'enzymes en utilisant la collagénase IA (2 mg/ml, Sigma Chemical Co., St. Louis, MO, USA) pendant une heure et la trypsine-EDTA (0.25%, GIBCO BRL, Gaithersbueg, MD, USA) pendant 20 minutes.During the myocardial infarction procedure, the anterior muscles of the right and left tibialis are dissected to separate the tendon and the aponeurotic tissue from the muscle tissue. They are then minced, weighed and dissociated with enzymes using collagenase IA (2 mg / ml, Sigma Chemical Co., St. Louis, MO, USA) for one hour and trypsin-EDTA (0.25%). , GIBCO BRL, Gaithersbueg, MD, USA) for 20 minutes.

Les cellules sont récoltées par sédimentation (7 min à 1200 rpm) et la réaction enzymatique est neutralisée par l'addition de 10% de sérum de veau foetal. Après passage sur un tamis de 100 µm et centrifugation, le surnageant est éliminé et les cellules sont resuspendues dans un milieu composé de F12(HAM) avec 20% de sérum de veau foetal, 1% (vol/vol) penicilline-streptomycine (10000 Ul/ml-10000 µg/ml, GIBCO BRL) et 5 ng/ml de bFGF (Sigma).The cells are harvested by sedimentation (7 min at 1200 rpm) and the enzymatic reaction is neutralized by the addition of 10% fetal calf serum. After passing through a 100 μm sieve and centrifugation, the supernatant is removed and the cells are resuspended in a medium composed of F12 (HAM) with 20% fetal calf serum, 1% (vol / vol) penicillin-streptomycin (10,000 IU / ml-10000 μg / ml, GIBCO BRL) and 5 μg / ml bFGF (Sigma).

L'ensemencement initial est réalisé en flacons de culture de 75 cm2 et les cellules sont incubées en air saturé en humidité à 5% de CO2.The initial seeding is carried out in culture flasks of 75 cm 2 and the cells are incubated in air saturated with moisture at 5% CO 2 .

Le jour de la transplantation, après 7 jours de culture et après évaluation fonctionnelle du niveau de base des fractions d'éjection ventriculaire par échocardiographie, les cellules ont été récoltées par trypsination, lavées et la viabilité a été testée. Un échantillon a été ensemencé sur des boîtes 12-puits dans 2.0 ml de milieu de culture pour comptage. Les cellules sont lavées dans le milieu d'injection (milieu de culture + BSA à 0.5%, Fraction V) et gardées dans la glace avant transplantation. Les cellules sont centrifugées, resuspendues dans 150 µl de milieu d'injection et administrées par voie sous-épicardique dans la zone infarcie.On day of transplantation, after 7 days of culture and after baseline functional evaluation of ventricular ejection fractions by echocardiography, the cells were harvested by trypsinization, washed and viability tested. A sample was seeded on 12-well dishes in 2.0 ml of culture medium for counting. The cells are washed in the injection medium (culture medium + 0.5% BSA, fraction V) and kept in the ice before transplantation. The cells are centrifuged, resuspended in 150 .mu.l of injection medium and administered sub-epicardially in the infarcted zone.

B.1.4 Transplantation des cellules dans la zone infarcieB.1.4 Transplantation of cells in the infarcted zone

Quarante-quatre rats ont fait partie de cette étude et ont été divisés en deux groupes : Un groupe contrôle et un groupe traité.Forty-four rats were included in this study and were divided into two groups: a control group and a treated group.

Tous les rats ont été réopérés une semaine après l'infarctus du myocarde, sous anesthésie générale et ventilation de la trachée. Tous les rats ont reçus 150 µl de milieu d'injection administré dans la zone infarcie au moyen d'une aiguille 30 Gauge. Dans le groupe contrôle (n=23), les rats ont reçu le milieu d'injection seul. Le groupe traité (n=21) a reçu la suspension de cellules myogéniques cultivées.All rats were reoperated one week after myocardial infarction, under general anesthesia and tracheal ventilation. All rats received 150 μl of injection medium administered in the infarcted zone using a 30-gauge needle. In the control group (n = 23), the rats received the injection medium alone. The treated group (n = 21) received the suspension of cultured myogenic cells.

Dans chaque groupe, quatre catégories de risques ont été étudiées par rapport à la fraction d'éjection de base LVEF : <25% (n=15), 25-35% (n=15) et >40% (n=16). Cette stratification permet d'obtenir des nombres homogènes d'animaux dans chaque sous-groupe afin de rendre les résultats statistiques plus précis.In each group, four risk categories were studied in relation to the LVEF baseline ejection fraction: <25% (n = 15), 25-35% (n = 15) and> 40% (n = 16) . This stratification makes it possible to obtain homogeneous numbers of animals in each subgroup in order to make the statistical results more precise.

B.1.5 Etudes immuno- et histochimiquesB.1.5 Immuno- and histochemical studies

Un jour après transplantation, les cellules ensemencées en boite de 12-puits sont fixées au méthanol et refroidies à -20°C pendant 5 minutes. Le marquage non spécifique est neutralisé en utilisant un mélange de 5% de sérum de cheval (HS) et 5% de sérum de veau foetal dans du PBS pendant 20 minutes. Les cellules sont incubées avec un anticorps de souris dirigé contre la desmine (1/200 DAKO, A/S-Denmark) pendant 1 heure puis avec un anticorps anti-souris conjugué au marqueur Cy3 (1/200, Jackson Immuno Research Laboratories, Inc.) pendant une heure dans l'obscurité.One day after transplantation, the cells seeded in a 12-well dish are fixed with methanol and cooled at -20 ° C. for 5 minutes. Non-specific staining is neutralized using a mixture of 5% horse serum (HS) and 5% fetal calf serum in PBS for 20 minutes. The cells are incubated with a mouse antibody directed against desmin (1/200 DAKO, A / S-Denmark) for 1 hour and then with an anti-mouse antibody conjugated to Cy3 marker (1/200, Jackson Immuno Research Laboratories, Inc. .) for an hour in the dark.

Les cellules sont observées à l'aide d'un microscope inversé à contraste de phase et à illumination fluorescente. Plusieurs clichés ont été pris de manière aléatoire. La proportion de myoblastes est calculée en divisant le nombre de cellules positives à la desmine par le nombre total de cellules observées.The cells are observed using an inverted phase contrast and fluorescent illumination microscope. Several snapshots were taken randomly. The proportion of myoblasts is calculated by dividing the number of desmin-positive cells by the total number of cells observed.

Dans les trois mois qui suivent la dernière échocardiographie (soit 2 mois après transplantation), les rats sont sacrifiés par une surdose de ketamine et de xylasine. Les ventricules sont isolés et sont sectionnés en deux parties le long de leur axe longitudinal. Les deux parties sont mises dans l'isopentane et congelées à l'azote. De fines sections de 8 µm sont préparées en utilisant un cryostat et des études classiques histologiques sont réalisées par coloration à l'hématoxyline et à l'éosine.Within three months after the last echocardiogram (ie 2 months after transplantation), the rats are sacrificed by an overdose of ketamine and xylasin. The ventricles are isolated and divided into two parts along their longitudinal axis. Both parts are put in isopentane and frozen with nitrogen. Thin 8 μm sections are prepared using a cryostat and standard histological studies are performed by hematoxylin and eosin staining.

B.1.6 Analyse statistiqueB.1.6 Statistical analysis

Toutes les données sont en moyenne à ± 1 SEM. Toutes les analyses ont été réalisées à l'aide du logiciel approprié (Statview 5.0, SAS Institute Inc., Cary, NC, USA). Le seuil critique α pour les analyses a été choisi à p<0.05.All data are on average ± 1 SEM. All assays were performed using the appropriate software (Statview 5.0, SAS Institute Inc., Cary, NC, USA). The critical threshold α for the analyzes was chosen at p <0.05.

Les comparaisons des variables continues entre les groupes contrôle et traité et chaque catégorie de risque ont été réalisées à l'aide de l'analyse de variance (méthode ANOVA) suivi du test post hoc (Scheffé). Les études longitudinales comparant les données échocardiographiques pour chaque groupe, avant et un et deux mois après l'injection intramyoacardique ont été réalisées en utilisant le test d'appariement.Comparisons of continuous variables between the control and treatment groups and each risk category were performed using analysis of variance (ANOVA method) followed by the post hoc test (Scheffé). Longitudinal studies comparing echocardiographic data for each group before and one and two months after intramyacardial injection were performed using the matching test.

Pour tester les relations entre le nombre de cellules injectées et la fonction cardiaque après transplantation, deux variables ont été créées : (LVEF à 1 mois/ LVEF) et (LVEF à 2 mois/LVEF). Le lien a été étudié par le calcul du F-ratio pour la régression ANOVA et le coefficient R2 ajusté pour les analyses de régression linéaire.To test the relationships between the number of cells injected and cardiac function after transplantation, two variables were created: (LVEF at 1 month / LVEF) and (LVEF at 2 months / LVEF). The link was studied by calculating the F-ratio for the ANOVA regression and the adjusted R 2 coefficient for the linear regression analyzes.

De plus, la variabilité à l'intérieur de chaque groupe des tests échocardiographiques a été observée à partir de deux lots de mesures effectuées sur 10 rats choisis de manière aléatoire en utilisant une analyse de Bland et Altman.In addition, the variability within each group of echocardiographic tests was observed from two batches of measurements on 10 randomly selected rats using Bland and Altman analysis.

B.2 Résultats B.2 Results B.2.1 Caractérisation de la suspension injectéeB.2.1 Characterization of the injected suspension

Sur 10000 cellules comptées le jour de la transplantation, 50% sont positives pour l'expression de la desmine. Le nombre de cellules injectées est de 3.500 000 ± 500000 s'étalant de 700000 à 6.5x106.Of the 10,000 cells counted on the day of transplantation, 50% are positive for the expression of desmin. The number of injected cells is 3,500,000 ± 500,000 ranging from 700,000 to 6.5x10 6 .

B.2.2 Test de fonctionnalité après transplantation des cellules d'origine musculaireB.2.2 Functional test after transplantation of muscle cells

Les paramètres échocardiographiques du niveau de base ne sont pas significativement différents entre les groupes. Au contraire, des différences majeures entre les groupes sont observées après la transplantation. Ainsi, dans le groupe traité, la fonction cardiaque est améliorée, comme cela est montré par comparaison de LVEF avec les groupes contrôles (Figure 1). A un mois après l'injection myocardique, des différences significatives sont observées entre les deux groupes (37.52 ± 1.92% contre 25.49 ± 2.47%, p=0.0005). Ce résultat est confirmé après 2 mois après l'injection (40.92 ± 2.17% contre 25.83 ± 2.39%, p<0.0001). Cette amélioration de LVEF dans le groupe traité est essentiellement reliée à une plus petite augmentation de LVESV par rapport à la dilatation ventriculaire, représentée par la variable LVEDV, qui augmente de manière similaire dans les deux groupes (Figure 2).The baseline echocardiographic parameters are not significantly different between the groups. On the contrary, major differences between groups are observed after transplantation. Thus, in the treated group, cardiac function is improved, as shown by comparison of LVEF with the control groups ( Figure 1 ). At one month after myocardial injection, significant differences were observed between the two groups (37.52 ± 1.92% vs. 25.49 ± 2.47%, p = 0.0005). This result is confirmed after 2 months after the injection (40.92 ± 2.17% against 25.83 ± 2.39%, p <0.0001). This improvement in LVEF in the treated group is essentially related to a smaller increase in LVESV compared to ventricular dilatation, represented by the variable LVEDV, which increases similarly in both groups ( Figure 2 ).

Les analyses longitudinales dans chaque groupe ont montré une amélioration substantielle de la fonction du ventricule gauche dans le groupe traité (Figure 3). Des différences significatives sont observées en comparant LVEF à un mois et LVEF à 2 mois à la variable LVEF du niveau de base. Les différences sont aussi significatives en comparant LVEF à un mois à LVEF à 2 mois. Tandis que LVEDV et LVESV sont augmentés à un mois en comparant aux valeurs de bases (p<0.0001 et p=0.0003, respectivement) et à 2 mois (p<0.0001 et p=0.029, respectivement), une stabilisation et une diminution sont montrées quand les valeurs à 2 mois sont comparées aux valeurs à 1 mois (p=0.78 et p=0.12, respectivement). Dans le groupe contrôle, une importante diminution de LVEF avec une augmentation significative de LVEDV est déjà visible à un mois (p=0.0066 et p<0.0001 contre valeurs de base respectivement). Les deux paramètres indiquent des valeurs similaires à 2 mois.Longitudinal analyzes in each group showed a substantial improvement in left ventricular function in the treated group ( Figure 3 ). Significant differences were observed comparing LVEF at one month and LVEF at 2 months at baseline LVEF variable. The differences are also significant when comparing LVEF at one month to LVEF at 2 months. While LVEDV and LVESV are increased to one month by comparing to baseline values (p <0.0001 and p = 0.0003, respectively) and at 2 months (p <0.0001 and p = 0.029, respectively), a stabilization and a decrease are shown when 2-month values are compared to 1-month values (p = 0.78 and p = 0.12, respectively). In the control group, a significant decrease in LVEF with a significant increase in LVEDV is already visible at one month (p = 0.0066 and p <0.0001 vs baseline respectively). Both parameters indicate values similar to 2 months.

Quand la fonction cardiaque (LVEF) est analysée par catégories de risques selon les valeurs de bases LVEF, des différences sont observées entre les groupes contrôle et traité à un mois dans les deux sous-groupes intermédiaires 25-35% et 35-40%. Cette amélioration de LVEF est confirmée à 2 mois après injection dans les deux sous-groupes, mais de manière intéressante, un effet bénéfique est aussi observé, en comparant le groupe traité au groupe contrôle dans le groupe de risque <25%.When cardiac function (LVEF) is analyzed by hazard categories according to LVEF baseline values, differences are observed between control and treated groups at one month in the two intermediate 25-35% and 35-40% subgroups. This improvement in LVEF is confirmed at 2 months after injection in both subgroups, but interestingly, a beneficial effect is also observed, comparing the treated group to the control group in the risk group <25%.

Enfin, l'étude de régression montre un lien significatif entre le nombre de myoblastes greffés et les ratios LVEF à un mois (R2=0.675, p<0.0001) et à 2 mois (R2=0.714, p<0.0001) (Figure 4). Quand les données sont analysées par groupe de risque, l'impact du nombre de cellules injectées à 2 mois est aussi significatif dans les sous-groupes <25%, 25-35% et 35-40% (R2=0.836, p=0.0106 ; R2=0.928, p=0.0083 et R2=0.985, p=0.0076, respectivement).Finally, the regression study shows a significant link between the number of grafted myoblasts and the LVEF ratios at one month (R 2 = 0.675, p <0.0001) and at 2 months (R 2 = 0.714, p <0.0001) ( Figure 4 ). When the data are analyzed by risk group, the impact of the number of cells injected at 2 months is also significant in the subgroups <25%, 25-35% and 35-40% (R 2 = 0.836, p = 0.0106, R 2 = 0.928, p = 0.0083 and R 2 = 0.985, p = 0.0076, respectively).

B.2.3 Effets cumulés de la transplantation des cellules d'origine musculaire et d'un traitement par un inhibiteur de l'enzyme de conversion de l'angiotensine (ACEI)B.2.3 Cumulative effects of muscle cell transplantation and treatment with an angiotensin converting enzyme (ACEI) inhibitor

La prise en charge de l'insuffisance cardiaque consiste à l'heure actuelle en l'administration d'inhibiteurs d'ACE. Il était par conséquent intéressant d'étudier s'il pouvait exister une synergie entre la transplantation de cellules d'origine musculaire obtenues selon le procédé de l'invention et l'effet protecteur procuré par les inhibiteurs d'ACE.The management of heart failure currently consists of the administration of ACE inhibitors. It was therefore interesting to study whether there could be a synergy between the transplantation of cells of muscle origin obtained according to the method of the invention and the protective effect provided by the ACE inhibitors.

Un infarctus du myocarde a été réalisé chez 39 rats par ligature des artères coronaires. Un traitement au perindoprilat à 1 mg/kg par jour, un inhibiteur de l'ACE, a été initié immédiatement après l'infarctus et continué sans interruption jusqu'au sacrifice de l'animal. Une semaine après l'infarctus, les animaux ont été opérés à nouveau et sélectionnés de manière aléatoire pour recevoir une injection sous-épicardique de 150 µl de milieu de culture seul (groupe contrôle, n=21) ou un volume équivalent contenant les cellules d'origine musculaire, soit environ 3x106 cellules cultivées à partir d'une biopsie de muscle de tibia antérieur et récoltées au moment de l'infarctus (groupe traité, n=18). La fonction ventriculaire gauche a été testée par échocardiographie un mois après la transplantation. Le niveau de base de la valeur de la fraction d'éjection est similaire entre les animaux contrôles (24 ± 2%) et les animaux traités (28 ± 1%) (p = 0.11). Un mois après la transplantation, les valeurs des fractions d'éjection ont augmenté dans les deux groupes et sont de 32% ± 2% pour le contrôle et de 38% ± 2% pour le groupe traité. Cependant, on constate une plus forte augmentation chez le groupe traité (p< 0.0001 contre niveau de base) par rapport au groupe contrôle (p=0.004 contre niveau de base), la valeur de fraction d'éjection étant significativement plus élevée chez les rats traités (p=0.04 contre le groupe contrôle). L'analyse des données de volume a montré que l'amélioration fonctionnelle apportée par la transplantation de cellules d'origine musculaire est essentiellement reliée à une augmentation de la contractilité plutôt qu'à une modification du ventricule gauche.Myocardial infarction was performed in 39 rats by ligation of the coronary arteries. Perindoprilat treatment at 1 mg / kg daily, an ACE inhibitor, was initiated immediately after infarction and continued without interruption until the animal was sacrificed. One week after the infarction, the animals were operated on again and randomly selected to receive a subepicardial injection of 150 μl of culture medium alone (control group, n = 21) or an equivalent volume containing the Muscle origin, approximately 3x10 6 cells cultured from anterior tibia muscle biopsy and harvested at the time of infarction (treated group, n = 18). Left ventricular function was tested by echocardiography one month after transplantation. The baseline ejection fraction value is similar between control animals (24 ± 2%) and treated animals (28 ± 1%) (p = 0.11). One month after transplantation, the values of ejection fractions increased in both groups and were 32% ± 2% for control and 38% ± 2% for the treated group. However, there was a greater increase in the treated group (p <0.0001 vs. baseline) compared to the control group (p = 0.004 vs. baseline), with the ejection fraction value being significantly higher in the rats. treated (p = 0.04 against the control group). Analysis of the volume data showed that the functional improvement provided by muscle-derived cell transplantation is primarily related to an increase in contractility rather than a change in the left ventricle.

Ces données montrent qu'il existe un effet additif de la transplantation de cellules d'origine musculaire et du traitement aux inhibiteurs d'ACE.These data show that there is an additive effect of muscle cell transplantation and treatment with ACE inhibitors.

C. ESSAIS CLINIQUES CHEZ L'HOMME DE TRANSPLANTATION DE CELLULES D'ORIGINE MUSCULAIRE POUR LA RECONSTITUTION DES TISSUS MYOCARDIQUESC. CLINICAL TRIALS IN MAN OF TRANSPLANTATION OF MUSCLE CELLS FOR THE RECONSTITUTION OF MYOCARDIAL TISSUES

Des essais cliniques de transplantations des cellules d'origine musculaire préparées selon le procédé de l'invention ont été réalisés chez l'homme en vue de traiter l'insuffisance cardiaque.Clinical trials of muscle-derived cell transplants prepared according to the method of the invention have been carried out in humans to treat heart failure.

C.1 Méthodes C.1 Methods

Les essais ont été réalisés chez 6 patients, dont, au moment de leur inclusion, l'âge était compris entre 18 et 75 ans. Ces patients ont tous fait l'objet d'indications cliniques de pontage aorto-coronaire avec possibilité technique de revascularisation chirurgicale. Leur fraction d'éjection globale ventriculaire gauche (mesurée par échocardiographie et/ou angiographie et/ou isotopes) était inférieure ou égale à 35%. Ils avaient tous des antécédents d'infarctus myocardique transmural. Enfin, les patients avait une hypo ou akinésie segmentaire touchant deux segments contigus ou plus (en dehors d'un anévrisme), en rapport avec l'infarctus. L'activité métabolique résiduelle dans cette zone était inférieure à 50%, détectée à la tomographie par émission de positons (TEP) et il n'y avait pas d'augmentation de cinétique à l'échographie Dobutamine à faible dose (10 gamma/Kg/mn).The trials were conducted in 6 patients, of whom, at the time of inclusion, the age was between 18 and 75 years. These patients have all had clinical indications of coronary artery bypass grafting with the technical possibility of surgical revascularization. Their left ventricular ejection fraction (measured by echocardiography and / or angiography and / or isotopes) was less than or equal to 35%. They all had a history of transmural myocardial infarction. Finally, the patients had segmental hypo or akinesia affecting two or more contiguous segments (outside an aneurysm), related to the infarct. Residual metabolic activity in this area was less than 50%, detected by positron emission tomography (PET) and there was no increase in kinetics on low dose Dobutamine ultrasound (10 gamma / kg / min).

Dix à dix huit grammes de tissus autologues sont prélevés au niveau du vastus lateralis chez le patient. L'incision est réalisée à l'aplomb du vaste externe. Le protocole de mise en culture et d'expansion des cellules est celui décrit dans la partie A du présent texte.Ten to eighteen grams of autologous tissue are collected from the vastus lateralis in the patient. The incision is made in line with the external vastus. The protocol for culturing and expanding the cells is that described in Part A of this text.

La culture cellulaire à injecter est ensuite déposée dans une cupule en inox stérile pour être aspirée dans une seringue de 1 ml. Un pontage coronaire est d'abord réalisé sous circulation extra-corporelle selon la technique habituelle. Après analyse de l'extension de l'infarctus et repérage des bords de la zone nécrosée, la suspension cellulaire d'environ 650 à 1200 millions de cellules (1,5x108 cellules/ml) est injectée, dans et aux confins de la zone infarcie, à l'aide de la seringue de 1 ml. Plusieurs injections sont nécessaires pour déposer la totalité des cellules. Ce temps opératoire est réalisé sous circulation extra-corporelle et clampage.The cell culture to be injected is then deposited in a sterile stainless steel cup to be sucked into a 1 ml syringe. A coronary bypass is first performed under extra-corporeal circulation according to the usual technique. After analysis of the extension of the infarct and identification of the edges of the necrotic zone, the cell suspension of approximately 650 to 1200 million cells (1.5x10 8 cells / ml) is injected into and at the confines of the zone. infarction, using the 1 ml syringe. Several injections are necessary to deposit all the cells. This operating time is performed under extracorporeal circulation and clamping.

C.2 Méthodes d'évaluations et résultats fonctionnels C.2 Evaluation methods and functional results

Ces essais ont permis de montrer la faisabilité et la sécurité de la technique chez l'homme. Par ailleurs, des tests de fonctionnalité sont réalisés par la mesure de la fonction ventriculaire gauche (segmentaire et globale) et du remodelage ventriculaire, couplée à la mesure de l'activité cellulaire métabolique. Ces mesures sont obtenues par des méthodes ultrasoniques telles que l'échocardiographie Doppler usuelle (mesure des diamètres, volumes et fractions d'éjection ventriculaire gauche), le Doppler tissulaire dans la zone infarcie et l'échocardiographie Dobutamine pour la recherche d'ischémie et de viabilité. Elles sont aussi obtenues par des méthodes isotopiques telles que la Tomographie à Emission de Positons (captation du désoxyfluoroglucose dans la zone infarcie).These tests have demonstrated the feasibility and safety of the technique in humans. In addition, functional tests are performed by measuring left ventricular function (segmental and global) and ventricular remodeling, coupled with the measurement of metabolic cellular activity. These measurements are obtained by ultrasonic methods such as the usual Doppler echocardiography (measurement of diameters, volumes and left ventricular ejection fractions), tissue Doppler in the infarcted zone and Dobutamine echocardiography for the investigation of ischemia and viability. They are also obtained by isotopic methods such as positron emission tomography (uptake of deoxyfluoroglucose in the infarcted zone).

Ces mesures sont réalisées en pré-opératoire et en post-opératoire au 1er mois ± 8 jours et au 3ème mois ± 8 jours.These measurements are performed preoperatively and postoperatively at 1 month ± 8 days and the 3 rd month ± 8 days.

Les résultats obtenus après transplantation de myoblastes sur un patient sont présentés ci-après.The results obtained after transplantation of myoblasts on a patient are presented below.

Le patient est un homme de 72 ans, hospitalisé pour insuffisance cardiaque (classe III de la NYHA), consécutive à un infarctus du myocarde inférieur étendu et réfractaire au traitement médical, ce d'autant que bêtabloquants et inhibiteurs de l'enzyme de conversion ont dû être interrompus, car non tolérés. Le bilan extracardiaque note par ailleurs une insuffisance rénale modérée (créatinine : 200 mmol/L) et une occlusion carotidienne bilatérale sans retentissement fonctionnel au doppler intracrânien et ne relevant par conséquent pas d'un geste vasculaire autonome.The patient is a 72-year-old man hospitalized for heart failure (NYHA Class III) following an extensive inferior infarction of myocardial infarction that is refractory to medical treatment, especially since beta-blockers and angiotensin-converting enzyme inhibitors have had to be interrupted because not tolerated. The extracardiac balance also notes a moderate renal insufficiency (creatinine: 200 mmol / L) and a bilateral carotid occlusion without functional repercussion to the intracranial Doppler and therefore not an autonomous vascular gesture.

A l'échocardiographie, la fraction d'éjection ventriculaire gauche est à 20% avec une akinésie étendue de la-paroi inférieure et une hypokinésie antéro-latérale sévère. L'absence de viabilité dans cette paroi inférieure est démontrée par la persistance de l'akinésie sous dobutamine à faible dose. La paroi antéro-latérale, en revanche, a une réponse biphasique à la dobutamine (faible dose, puis forte dose) témoignant d'une viabilité et d'une ischémie. Ces données sont confirmées par la tomographie d'émission de positrons (TEP) au 2fluoro18désoxyglucose (FDG) qui montre que la viabilité fait totalement défaut dans la paroi inférieure, mais qu'elle est présente dans les portions antérieure et latérale du ventricule gauche.On echocardiography, the left ventricular ejection fraction is 20% with extensive akinesia of the lower wall and severe anterolateral hypokinesia. The lack of viability in this lower wall is demonstrated by the persistence of akinesia under low dose dobutamine. The anterolateral wall, on the other hand, has a biphasic response to dobutamine (low dose, then high dose) indicating viability and ischemia. These data are confirmed by positron emission tomography (PET) with 2 fluoro 18 deoxyglucose (FDG), which shows that viability is totally lacking in the lower wall, but is present in the anterior and lateral portions of the ventricle. left.

Sur la coronarographie, il existe une occlusion complète, proximale, de l'artère interventriculaire antérieure, avec une opacification tardive du lit d'aval par des collatérales homolatérales, une sténose serrée d'une branche diagonale haute et une occlusion proximale de l'artère coronaire droite. L'artère coronaire droite n'a que des irrégularités non significatives. Le présent patient présente donc en résumé :

  1. (1) une altération importante de la fonction ventriculaire gauche,
  2. (2) une cicatrice d'infarctus akinétique et métaboliquement non viable,
  3. (3) une indication élective de pontage sur les artères autres que celles de l'infarctus.
On the coronarography, there is a complete, proximal occlusion of the anterior interventricular artery, with a late opacification of the downstream bed by ipsilateral collaterals, a tight stenosis of high diagonal branch and proximal occlusion of the right coronary artery. The right coronary artery has only insignificant irregularities. The present patient summarizes:
  1. (1) significant impairment of left ventricular function,
  2. (2) an akinetic and metabolically unsustainable myocardial infarction scar,
  3. (3) an elective indication of bypass on the arteries other than those of the infarct.

Sous anesthésie locale, un fragment du vaste externe est prélevé du patient au travers d'une courte incision (5 cm). Les myoblastes sont produits selon le procédé de l'invention décrit en partie A. Le nombre de cellules est multiplié à l'aide de plusieurs expansions en plateaux multi-étagés permettant d'obtenir en deux semaines 800x106 cellules, dont 65% de cellules CD56+. La proportion de cellules viables est supérieure à 96%.Under local anesthesia, a fragment of the vastus externus is removed from the patient through a short incision (5 cm). The myoblasts are produced according to the method of the invention described in part A. The number of cells is multiplied using several expansions in multi-stage trays to obtain in two weeks 800x10 6 cells, 65% of cells CD56 +. The proportion of viable cells is greater than 96%.

Deux semaines après la biopsie, le patient est réhospitalisé dans le service de chirurgie cardiaque, en vue de son pontage. Compte tenu de la précarité de l'état hémodynamique après l'induction anesthésique (débit cardiaque à 1,5 Umin avec une saturation veineuse en oxygène à 55%) une contrepulsion par ballon intraaortique est instaurée de façon prophylactique. Les artères diagonale et interventriculaire antérieures sont pontées à l'aide d'un greffon veineux saphène et de l'artère mammaire interne gauche, respectivement, sous circulation extracorporelle et cardioplégie sanguine chaude, rétrograde, continue. Après réalisation des anastosomes coronaires, la zone infarcie de la paroi inférieure est aisément identifiée. Trente-trois injections de cellules, suspendues dans 5 ml d'albumine, sont faites dans et autour des foyers blanchâtres de nécrose à l'aide d'une aiguille de 27 G à angle droit spécialement conçue pour permettre la création de chenaux sous-épicardiques d'aspect pratiquement phlycténulaire. La durée du clampage aortique est de 56 min, dont 16 consacrées aux injections des cellules. Il n'y a aucun saignement à partir des sites d'injection, et la circulation extracorporelle peut être arrêtée sans difficultés. Le patient est sevré de la contrepulsion et d'un support pharmacologique par dobutamine au cours des 3 premiers jours postopératoires et sort au 8e jour après des suites simples.Two weeks after the biopsy, the patient is readmitted to the cardiac surgery department for bridging. Given the precariousness of the hemodynamic state after anesthetic induction (cardiac output at 1.5 Umin with a 55% oxygen venous saturation) an intraaortic balloon counterpulsation is instituted prophylactically. The anterior diagonal and interventricular arteries are bridged with a saphenous vein graft and the left internal mammary artery, respectively, under extracorporeal circulation and warm, retrograde, continuous blood cardioplegia. After completion of the coronary anastosomes, the infarcted area of the lower wall is easily identified. Thirty-three cell injections, suspended in 5 ml of albumin, are made in and around whitish foci of necrosis using a right-angle 27 G needle specially designed to allow the creation of subepicardial channels. almost phlyctenular in appearance. The duration of aortic cross-clamping is 56 min, of which 16 are dedicated to cell injections. There is no bleeding from injection sites, and extracorporeal circulation can be stopped without difficulty. The patient is weaned from the counterpulsation and pharmacological support with dobutamine in postoperative first 3 days and leaves the 8th day after uneventful.

Huit mois plus tard, son état clinique s'est amélioré, et il est actuellement en classe Il de NYHA, alors que le traitement médical est resté inchangé. Un Holter des 24 heures ne montre aucun trouble du rythme. Les études de 4 échocardiographies effectuées chaque mois depuis l'opération chirurgicale montrent que la fraction d'éjection du ventricule gauche a augmenté à 30%. Comme le montre la figure 5, la contractilité segmentaire est démontrée non seulement dans la paroi antérieure, mais aussi dans la zone postérieure infarcie et greffée qui se contracte (le pourcentage d'épaississement systolique est passé de valeurs préopératoires quasiment indétectable à 40%.Eight months later, his clinical condition improved, and he is currently in NYHA Class II, while medical treatment has remained unchanged. A 24-hour Holter shows no disturbance of rhythm. Studies of 4 echocardiograms performed each month since the surgical operation show that the left ventricular ejection fraction has increased to 30%. As shown in figure 5 Segmental contractility is demonstrated not only in the anterior wall, but also in the contracting infarcted and grafted posterior zone (the percentage of systolic thickening has gone from preoperative values almost undetectable to 40%).

Fait nouveau, cette contractilité s'améliore encore sous dobutamine. Par ailleurs, une imagerie doppler tissulaire montre l'apparition d'un gradient de vélocité transmyocardique en systole. Une nouvelle tomographie à émission de positrons au FDG montre clairement la captation du traceur dans la paroi inférieure, avec un ratio d'activité entre la paroi et le septum (pris comme contrôle), qui augmente de 0,5 avant l'opération à 0,7, ce qui reflète une nouvelle activité métabolique dans la zone infarcie dépourvue de viabilité en préopératoire (Figure 6). Cette dernière observation ne peut avoir été influencée par la revascularisation myocardique associée et, confrontée aux données échocardiographiques, suggère que l'amélioration fonctionnelle dans la zone infarcie est réellement en rapport avec la présence des myoblastes greffés.Again, this contractility is still improving under dobutamine. In addition, tissue Doppler imaging shows the appearance of a transmyocardial velocity gradient in systole. A new FDG positron emission tomography clearly shows the uptake of the tracer in the lower wall, with a ratio of activity between wall and septum (taken as a control), which increases from 0.5 before the operation to 0 , 7, which reflects a new metabolic activity in the infarcted zone devoid of preoperative viability ( Figure 6 ). This last observation can not have been influenced by the associated myocardial revascularization and, confronted with the echocardiographic data, suggests that the functional improvement in the infarcted zone is really related to the presence of grafted myoblasts.

Pris dans leur ensemble, ces résultats montrent que la fonction de la zone infarcie a été améliorée par la transplantation de myoblastes obtenus selon le procédé de l'invention.Taken together, these results show that the function of the infarcted zone was improved by the transplantation of myoblasts obtained according to the method of the invention.

L'injection de cellules musculaires autologues, préparées selon le procédé, a encore été réalisée sur 5 autres patients. Les tableaux suivants résument les données du suivi clinique sur 4 des 5 patients (nommés respectivement MYO3, MYO4, MYO5 et MYO6). Le suivi clinique du sixième patient est en cours. Nom du patient D. MYO 3 Age 62 Sexe M Type de Pathologie Insuffisance cardiaque post-ischémique, angor instable Fraction d'éjection 25 % Stade classification NYHA III PET-Scan Présence d'une zone non viable Échographie initiale Akinésie antérieure, dyskinésie apicale, hypokinésie latérale Bilan virologique Négatif Durée de culture 18 J Cellules : nombre, caractéristiques 922 x 106. CD56+ : 91 % ; CD15+ : 6 % ; Desmine+ : 65 % ; Viabilité : 98 %; Formation de myotubes (test fonctionnel) : +. Siège d'injection Antérieure ; 900 x 106 cellules injectées Complications infectieuses Absence complications infectieuses post-opératoires Évaluation fonctionnelle :

  • Stade NYHA
  • Fraction d'éjection
  • Contractilité segmentaire
  • III -> II
  • 25 % -> 35%
  • Augmentation (+/-)
Évolution viabilité segmentaire Amélioration (+) Nom du patient E. MYO 4 Age 67 Sexe M Type de Pathologie Insuffisance cardiaque post-ischémique Fraction d'éjection 31 % Stade classification NYHA III PET-Scan Présence d'une zone non viable Échographie initiale Akinésie apicale, akinésie postérieure, hypokinésie latérale Bilan virologique Négatif Durée de culture 20 J Cellules : nombre, caractéristiques 657 x 106. CD56+ : 97 % ; CD15+ : 15 % ; Desmine+ : 58 % ; HLA Classel : 94 % ; Viabilité : 98 %; Formation de myotubes (test fonctionnel) : +. Siège d'injection Postérieure ; 620 x 106 cellules injectées Complications infectieuses Absence complications infectieuses post-opératoires Évaluation fonctionnelle :
  • Stade NYHA
  • Fraction d'éjection
  • Contractilité segmentaire
  • III -> II
  • 31 % -> 50%
  • Augmentation (+/-)
Évolution viabilité segmentaire Amélioration (+)
Nom du patient F. MYO 5 Age 39 Sexe M Type de Pathologie Insuffisance cardiaque post-ischémique Fraction d'éjection 22 % Stade classification NYHA III PET-Scan Présence d'une zone non viable Échographie initiale Akinésie apicale, akinésie postérieure, hypokinésie antérieure, hypokinésie latérale Bilan virologique Négatif Durée de culture 14 J Cellules : nombre, caractéristiques 993 x 106. CD56+ : 95 % ; CD15+ : 4 % ; Desmine+ : 78 % ; HLA Classel : 96 % ; Viabilité : 98 %; Formation de myotubes (test fonctionnel) : +. Siège d'injection Postéro-latérale ; 950 x 106 cellules injectées Complications infectieuses Absence complications infectieuses post-opératoires Évaluation fonctionnelle :
  • Stade NYHA
  • Fraction d'éjection
  • Contractilité segmentaire
  • III -> II
  • 22 % -> 36%
  • Augmentation (+)
Évolution viabilité segmentaire Amélioration (+)
Nom du patient G. MYO 6 Age 55 Sexe M Type de Pathologie Insuffisance cardiaque post-ischémique Fraction d'éjection 34 % Stade classification NYHA IV PET-Scan Présence d'une zone non viable Échographie initiale Akinésie antérieure, akinésie latérale, dyskinésie apicale, hypokinésie septale Bilan virologique Négatif Durée de culture 16 J Cellules : nombre, caractéristiques 1210 x 106. CD56+ : 85 % ; CD15+ : 10 % ; Desmine+ : 85 % ; HLA Classel : 96 % ; Viabilité : 97 %; Formation de myotubes (test fonctionnel) : +. Siège d'injection Antérieure ; 1150 x 106 cellules injectées Complications infectieuses Avant injection, patient en état de choc cardiovasculaire, sous adrénaline et noradrénaline. Absence complications infectieuses post-opératoires (24 h) après injection. Évaluation fonctionnelle : Patient décédé le 28/04/01. Cause du décès non imputable à l'injection cellulaire (choc cardio-vasculaire suivi d'ischémie mésentérique probables)
  • Stade NYHA
  • Fraction d'éjection
  • Contractilité segmentaire
Évolution viabilité segmentaire Sans objet
The injection of autologous muscle cells, prepared according to the method, was again performed on 5 other patients. The following tables summarize clinical follow-up data for 4 of 5 patients (named MYO3, MYO4, MYO5 and MYO6, respectively). The clinical follow-up of the sixth patient is in progress. Patient's name D. MYO 3 Age 62 Sex M Type of Pathology Post-ischemic heart failure, unstable angina Ejection fraction 25% NYHA classification stage III PET Scan Presence of a non-viable area Initial ultrasound Anterior ankinesia, apical dyskinesia, lateral hypokinesia Virological assessment Negative Culture time 18 J Cells: number, characteristics 922 x 10 6 . CD56 +: 91%; CD15 +: 6%; Desmin +: 65%; Viability: 98%; Formation of myotubes (functional test): +. Injection seat Anterior; 900 x 10 6 cells injected Infectious complications Absence of postoperative infectious complications Functional assessment:
  • NYHA Stadium
  • Ejection fraction
  • Segmental contractility
  • III -> II
  • 25% -> 35%
  • Increase (+/-)
Evolution segmental viability Improvement (+)
Patient's name E. MYO 4 Age 67 Sex M Type of Pathology Post-Ischemic Heart Failure Ejection fraction 31% NYHA classification stage III PET Scan Presence of a non-viable area Initial ultrasound Apical akinesia, posterior akinesis, lateral hypokinesia Virological assessment Negative Culture time 20 J Cells: number, characteristics 657 x 10 6 . CD56 +: 97%; CD15 +: 15%; Desmin +: 58%; HLA Classel: 94%; Viability: 98%; Formation of myotubes (functional test): +. Injection seat Posterior; 620 x 10 6 cells injected Infectious complications Absence of postoperative infectious complications Functional assessment:
  • NYHA Stadium
  • Ejection fraction
  • Segmental contractility
  • III -> II
  • 31% -> 50%
  • Increase (+/-)
Evolution segmental viability Improvement (+)
Patient's name F. MYO 5 Age 39 Sex M Type of Pathology Post-Ischemic Heart Failure Ejection fraction 22% NYHA classification stage III PET Scan Presence of a non-viable area Initial ultrasound Apical akinesia, posterior akinesia, anterior hypokinesia, lateral hypokinesia Virological assessment Negative Culture time 14 J Cells: number, characteristics 993 x 10 6 . CD56 +: 95%; CD15 +: 4%; Desmin +: 78%; HLA Classel: 96%; Viability: 98%; Formation of myotubes (functional test): +. Injection seat Postero-lateral; 950 x 10 6 cells injected Infectious complications Absence of postoperative infectious complications Functional assessment:
  • NYHA Stadium
  • Ejection fraction
  • Segmental contractility
  • III -> II
  • 22% -> 36%
  • Increase (+)
Evolution segmental viability Improvement (+)
Patient's name G. MYO 6 Age 55 Sex M Type of Pathology Post-Ischemic Heart Failure Ejection fraction 34% NYHA classification stage IV PET Scan Presence of a non-viable area Initial ultrasound Anterior ankinesia, lateral akinesia, apical dyskinesia, septal hypokinesia Virological assessment Negative Culture time 16 J Cells: number, characteristics 1210 x 10 6 . CD56 +: 85%; CD15 +: 10%; Desmin +: 85%; HLA Classel: 96%; Viability: 97%; Formation of myotubes (functional test): +. Injection seat Anterior; 1150 x 10 6 cells injected Infectious complications Before injection, patient with cardiovascular shock, adrenaline and norepinephrine. Absence of postoperative infectious complications (24 h) after injection. Functional assessment: Patient died on 28/04/01. Cause of death not attributable to cellular injection (cardiovascular shock followed by probable mesenteric ischemia)
  • NYHA Stadium
  • Ejection fraction
  • Segmental contractility
Evolution segmental viability Not applicable

En résumé, ces essais ont montré la possibilité d'obtenir le nombre de cellules souhaité dans un délai limité de 2 à 4 semaines. Ils ont par ailleurs montré que le prélèvement, la préparation et la transplantation des cellules musculaires autologues humaines pouvaient être réalisés selon l'invention sans difficultés et complications pré, per ou post-chirurgicales.In summary, these tests showed the possibility of obtaining the desired number of cells within a limited time of 2 to 4 weeks. They have also shown that the collection, preparation and transplantation of human autologous muscle cells could be carried out according to the invention without difficulties and pre- or post-surgical complications.

Enfin, ces essais ont de plus permis d'observer une amélioration clinique, une augmentation de la contractilité régionale (échographie) associée à une augmentation de la surface de zone viable (PET-Scan) chez ces patients.Finally, these trials have also shown a clinical improvement, an increase in regional contractility (ultrasound) associated with an increase in the viable area area (PET-Scan) in these patients.

En conclusion, les études présentées dans les parties B et C permettent d'apporter les différents résultats suivants :

  1. a) elles montrent que la transplantation des cellules d'origine musculaire améliore de manière significative la fonction ventriculaire gauche suite à un infarctus du myocarde,
  2. b) que l'amélioration est clairement dépendante du nombre de cellules injectées,
  3. c) que cette transplantation potentialise un traitement pharmacologique, notamment par une inhibition de l'ACE.
  4. d) Et enfin que le procédé de l'invention est adapté pour le traitement de l'insuffisance cardiaque chez l'homme.
  1. 1. Emerich, D.F. (1993). Cell transplantations of Huntington's disease. Cell transplantation 4: 348 .
  2. 2. Borlongan, C.V. and Sandberg, P. (1993). Microtransplantation of nigral dopamine neurons in a rat model of Parkinson's disease. Cell Transplantation 4: 347 .
  3. 3. Hering, B.J., Browatzki, C.C., Schultz, A., Bretzel, R.G. and Federlin, K.F. (1993). Clinical islet transplantation - registry report, accomplishments in the past and future research needs. Cell Transplantation 4: 269 .
  4. 4. Tremblay JP, Malouin F, Roy R, Huard J, Bouchard JP, Satoh A, Richards CL. (1993). Results of a triple blind clinical study of myoblast transplantations without immunosuppressive treatment in young boys with Duchenne muscular dystrophy. Cell Transplant 2: 99-112 .
  5. 5. Skuk D, Roy B, Goulet M, Tremblay JP. Successful myoblast transplantation in primates depends on appropriate cell delivery and induction of régénération in the host muscle. (1999) Exp. Neuro/ 155: 22-30
  6. 6. Law PK, Bertorini TE, Goodwin TG, Chen M, Fang Q, Li H-J, Kirby DS, Florendo JA, Herrod HG, Golden GS. (1990) Dystrophin production induced by myoblast transfer therapy in Duchenne muscular dystrophy. Lancet 336: 114-115 .
  7. 7. Huard J, Bouchard JP, Roy R, Malouin F, Dansereau G, Labrecque C, Albert N, Richards CL, Lemieux B, Tremblay JP. (1992). Human myoblast transplantation : preliminary results of 4 cases. Muscle Nerve 15: 550-560 .
  8. 8. Dhawan, J., Pan, L.C., Pavlath, O.K., Travis, M.A., Lanctot, A.M. and Blau, H.M. (1991). Systemic delivery of human growth hormone by injection of genetically engineered myoblasts. Science 254: 1509-1512 .
  9. 9. Dai, Y., Roman, M., Navlaux, R.K. and Verma, I.M. (1992). Gene therapy via primary myoblasts: long-term expression of factor IX protein following transplantation in vivo. Proc. Natl. Acad. Sci. USA 89: 10892-10895 .
  10. 10. Jiao, S, Gurevich, V. Wolff, J.A. (1993). Long-term correction of rat model of Parkinson's disease by gene therapy. Nature 362: 450-453
  11. 11. Gussoni, E, Blau, HM, Kunkel, LM. (1997). The fate of individual myoblasts after transplantation into muscles of DMD patients. Nature Medicine 3:970-977 .
  12. 12. Murry CE, Wiseman RW, Schwartz SM and Hauschka SD. (1996) Skeletal myoblast transplantation for repair of myocardial necrosis. J. Clin Invest 98: 2512-2523 .
  13. 13. Taylor, D.A., Atkins, B.Z., Hungspreugs, P., Jones, T.R., Reedy, M.C. et al. (1998). Regenerating functional myocardium: improved performance after skeletal myoblast transplantation. Nat. Med. 4: 929-33 .
  14. 14. Chiu, R.C.-J., Zibaitis, A. and Kao R.L. (1995). Cellular cardiomyoplasty: myocardial régénération with satellite cell implantation. Ann. Thorac. Surg. 60: 12-18 .
  15. 15. Gussoni E, Soneoka Y, Strickland CD, Buzney EA, Khan MK, Flint AF, Kunkel LM, Mulligan RC. (1999). Dystrophin expression in the mdx mouse restored by stem cell transplantation. Nature 401:390-394 .
  16. 16. Jackson KA, Mi T, Goodell MA. Hematopoietic potential of stem cells isolated from murine skeletal muscle. (1999) Proc Natl Acad Sci USA 96:14482-14486 .
  17. 17. Young, H.E., Steele, T.A., Bray, R.A., Detmer, K., Blake, L.W., Lucas, P.W., Black, A.S. (1999). Human pluripotent and progenitor cells display cell surface cluster differenciation markers CD10, CD13, CD56, and MHC Class-1. Proc Soc Exp Biol Med 221: 63-71
  18. 18. PCT WO 98/54301 (Mickle, D.A., Weisel, R. ). Transplants for myocardial scars and method and cellular preparations therefor.
  19. 19. PCT WO 96/18303 (Law, P.K. ). Myoblast therapy for mammalian disease.
  20. 20. EP 0 898 967 A1 (Law, P.K. ). Myoblast transfer therapy for relieving pain and for treating behavioural and perceptive abnormalities.
  21. 21. Webster, C., Pavlath, G.K., Parks, D.R., Walsh, F.S. and Blau, H.M. (1988). Isolation of human myoblasts with the fluorescence-activated cell sorter. Exp Cell Res 174: 252-265 .
  22. 22. Pittenger, M.F., Mackay, A.M., Beck, S.C., Jaiswal, R.K., Douglas, R., Mosca, J.D., Moorman, R.K., Simonetti, D.W., Craig, S., and Marshak, D.R. (1999). Multilineage potential of adult human mesenchymal stem cells. Science 284: 143 -
  23. 23. Ham RG, StClair JA, Webster C, Blau HM. (1988) Improved media for normal human muscle satellite cells: serum-free clonal growth and enhanced growth with low serum. In Vitro Cell Dev Biol 24: 833-844 .
  24. 24. Robinson S.W., Cho, P.C., Levitsky, H.I., Olson, J.L., Hruban, R.H., et al. (1996). Arterial delivery of genetically labelled skeletal myoblasts to the murine heart: long-term survival and phenotypic modification of implanted myoblasts. Cell Transplant 5: 77-91 .
  25. 25. Zhuquing Qu et al. Development of approaches to improve cell survival in myoblast transfer therapy. J. Cell Biol. 142: 1257-1267 .
  26. 26. Lequerica et al. In vitro proliferation, differentiation and immunomagnetic bead purification of human myoblast. Annals of transplantation (1999) 4: 103-108
  27. 27. WO 99/56785 (Université Pittsburg, 1999-11-11).
In conclusion, the studies presented in parts B and C allow to bring the following different results:
  1. a) they show that transplantation of muscle-derived cells significantly improves left ventricular function following myocardial infarction,
  2. b) that the improvement is clearly dependent on the number of cells injected,
  3. c) that this transplantation potentiates a pharmacological treatment, in particular by inhibition of ACE.
  4. d) And finally that the method of the invention is suitable for the treatment of heart failure in humans.
  1. 1. Emerich, DF (1993). Cell transplants of Huntington's disease. Cell transplantation 4: 348 .
  2. 2. Borlongan, CV and Sandberg, P. (1993). Microtransplantation of nigral dopamine neurons in a rat model of Parkinson's disease. Cell Transplantation 4: 347 .
  3. 3. Hering, BJ, Browatzki, CC, Schultz, A., Bretzel, RG and Federlin, KF (1993). Clinical islet transplantation - registry report, accomplishments in the past and future research needs. Cell Transplantation 4: 269 .
  4. 4. Tremblay JP, Malouin F, Roy R, Huard J, JP Bouchard, Satoh A, Richards CL. (1993). Results of a triple blind clinical study of myoblast transplants without immunosuppressive treatment in young boys with Duchenne muscular dystrophy. Cell Transplant 2: 99-112 .
  5. 5. Skuk D, Roy B, Goulet M, JP Tremblay. Successful myoblast transplantation in primates depends on appropriate delivery and induction of regeneration in the host muscle. (1999) Exp. Neuro / 155: 22-30
  6. 6. Law PK, Bertorini TE, TG Goodwin, Chen M, Fang Q, HJ Li, Kirby DS, Florendo JA, Herrod HG, Golden GS. (1990) Dystrophin production induced by myoblast transfer therapy in Duchenne muscular dystrophy. Lancet 336: 114-115 .
  7. 7. Huard J, JP Bouchard, Roy R, Malouin F, Dansereau G, Labrecque C, Albert N, CL Richards, Lemieux B, JP Tremblay. (1992). Human myoblast transplantation: preliminary results of 4 cases. Muscle Nerve 15: 550-560 .
  8. 8. Dhawan, J., Pan, LC, Pavlath, OK, Travis, MA, Lanctot, AM and Blau, HM (1991). Systemic delivery of human growth hormone by injection of genetically engineered myoblasts. Science 254: 1509-1512 .
  9. 9. Dai, Y., Roman, M., Navlaux, RK and Verma, IM (1992). Gene therapy via primary myoblasts: long-term expression of factor IX protein following in vivo transplantation. Proc. Natl. Acad. Sci. USA 89: 10892-10895 .
  10. 10. Jiao, S, Gurevich, V. Wolff, JA (1993). Long-term correction of Parkinson's disease model by gene therapy. Nature 362: 450-453
  11. 11. Gussoni, E, Blau, HM, Kunkel, LM. (1997). The fate of individual myoblasts after transplantation into the muscles of DMD patients. Nature Medicine 3: 970-977 .
  12. 12. Murry CE, RW Wiseman, SM Schwartz and Hauschka SD. (1996) Skeletal myoblast transplantation for repair of myocardial necrosis. J. Clin Invest 98: 2512-2523 .
  13. 13. Taylor, DA, Atkins, BZ, Hungspreugs, P., Jones, TR, Reedy, MC et al. (1998). Regenerating functional myocardium: improved performance after skeletal myoblast transplantation. Nat. Med. 4: 929-33 .
  14. 14. Chiu, RC-J., Zibaitis, A. and Kao RL (1995). Cellular cardiomyoplasty: myocardial regeneration with satellite cell implantation. Ann. Thorac. Surg. 60: 12-18 .
  15. 15. Gussoni E, Soneoka Y, CD Strickland, EA Buzney, MK Khan, Flint AF, LM Kunkel, Mulligan RC. (1999). Dystrophin expression in the mdx mouse restored by stem cell transplantation. Nature 401: 390-394 .
  16. 16. Jackson KA, Mi T, Goodell MA. Hematopoietic potential of stem cells isolated from murine skeletal muscle. (1999) Proc Natl Acad Sci USA 96: 14482-14486 .
  17. 17. Young, HE, Steele, TA, Bray, RA, Detmer, K., Blake, LW, Lucas, PW, Black, AS (1999). Human pluripotent and progenitor cells display cell surface cluster differentiation markers CD10, CD13, CD56, and MHC Class-1. Proc Soc Exp Biol Med 221: 63-71
  18. 18. PCT WO 98/54301 (Mickle, DA, Weisel, R. ). Transplants for myocardial scars and method and cellular preparations therefor.
  19. 19. PCT WO 96/18303 (Law, PK ). Myoblast therapy for mammalian disease.
  20. 20. EP 0 898 967 A1 (Law, PK ). Myoblast transfer therapy for relieving pain and for treating behavioral and perceptive abnormalities.
  21. 21. Webster, C., Pavlath, GK, Parks, DR, Walsh, FS and Blau, HM (1988). Isolation of human myoblasts with fluorescence-activated cell sorter. Exp Cell Res 174: 252-265 .
  22. 22. Pittenger, MF, Mackay, AM, Beck, SC, Jaiswal, RK, Douglas, R., Mosca, JD, Moorman, RK, Simonetti, DW, Craig, S., and Marshak, DR (1999). Multilineage potential of adult human mesenchymal stem cells. Science 284: 143 -
  23. 23. Ham RG, StClair JA, Webster C, Blau HM. (1988) Improved media for normal human muscle satellite cells: serum-free clonal growth and enhanced growth with low serum. In Vitro Cell Dev Biol 24: 833-844 .
  24. 24. Robinson SW, Cho, PC, Levitsky, HI, Olson, JL, Hruban, RH, et al. (1996). Arterial delivery of genetically labeled skeletal myoblasts to the murine heart: long-term survival and phenotypic modification of implanted myoblasts. Cell Transplant 5: 77-91 .
  25. 25. Zhuquing Qu et al. In the field of myoblast transfer therapy. J. Cell Biol. 142: 1257-1267 .
  26. 26. Lequerica et al. In vitro proliferation, differentiation and immunomagnetic purification of human myoblast. Annals of transplantation (1999) 4: 103-108
  27. 27. WO 99/56785 (Pittsburg University, 1999-11-11).

Claims (23)

  1. A method for preparing a composition intended for cell therapy in humans, said method comprising the following steps:
    a) mincing a skeletal muscle tissue biopsy;
    b) enzymatic dissociation of the muscle fibres and cells and separating the individualized cells by filtering;
    c) culturing the cells obtained in step b) in a reactor for culturing adherent cells in the presence of a medium comprising MCDB 120 and D-valine substituted for L-valine until a population of cells comprising CD56+ cells as the dominant cell type is obtained, said cell type being dominant when the proportion of this cell type in said cell population exceeds 50%, said culture if necessary comprising one or more expansion phases;
    d) harvesting the cell population obtained in step c);
    e) if necessary, freezing the cell population harvested in step d).
  2. A method according to claim 1, characterized in that the harvested cell population comprises 50 x 106 cells to 800 x 109 cells, preferably at least 500 x 106 cells, among which myoblast cells expressing the CD56 marker constitute said dominant cell type.
  3. A method according to claim 2, characterized in that the harvested cell population comprises at least 50%, preferably 60%, more preferably 70% of myoblast cells expressing the CD56 marker.
  4. A method according to claim 2 or claim 3, characterized in that said medium contains a glucocorticoid and bFGF.
  5. A method according to any one of claims 2 to 4, characterized in that the composition is intended for reconstituting skeletal, cardiac or visceral muscle tissue in humans.
  6. A method according to any one of claims 2 to 5, characterized in that the composition is intended for the treatment of post-ischemic cardiac insufficiency in humans.
  7. A method according to any one of claims 2 to 6, characterized in that the composition is intended for the treatment of innate or acquired muscular dystrophy.
  8. A method according to any one of claims 2 to 7, characterized in that said mincing step is assisted by mechanical or electrical blades.
  9. A method according to any one of claims 2 to 8, characterized in that it further comprises a step for CD34+ cell depletion.
  10. A cell therapy product for its use in the treatment of a cardiac pathology in humans, comprising a population of 500 x 106 to 800 x 109 cells, said population comprising at least 50% of CD56+ cells.
  11. A cell therapy product according to claim 10, characterized in that said population comprises at least 60% CD56+ cells.
  12. A cell therapy product according to claim 10, characterized in that said population comprises at least 70% CD56+ cells.
  13. A cell therapy product according to any one of claims 10 to 12, characterized in that said treatment of a cardiac pathology is a treatment by repair of cardiac tissue in humans.
  14. A cell therapy product according to any one of claims 10 to 13, characterized in that said cardiac pathology is post-ischemic cardiac insufficiency.
  15. A cell therapy product according to claim 14, characterized in that said post-ischemic cardiac insufficiency treatment is carried out concomitantly with a treatment by an angiotensin conversion enzyme inhibitor (ACEI).
  16. A cell therapy product according to any one of claims 10 to 15, characterized in that said treatment of a cardiac pathology is a treatment by autologous transplantation.
  17. Use of a cell population as defined in any one of claims 10 to 16, for the production of a cell therapy product intended for the treatment of a cardiac pathology.
  18. A culture medium comprising MCDB 120 and D-valine substituted for L-valine.
  19. A culture medium according to claim 18, which does not comprise phenol red and which does not comprise thymidine.
  20. In vitro use of a culture medium according to claim 18 or claim 19, for the differentiation of skeletal muscle tissue cells into myoblasts.
  21. In vitro use of a culture medium according to claim 18 or claim 19, for the production of a cell population comprising a dominant cell type selected from CD34+ cells, CD15+ cells, CD56+ cells, HLA class 1 cells, said cell type being dominant when the proportion of this cell type exceeds 50%.
  22. Use according to claim 21, characterized in that said dominant cell type is selected from CD34+ cells, CD15+ cells and CD56+ cells.
  23. In vitro use of a culture medium according to claim 18 or claim 19, for the production of a cell therapy product comprising a cell population comprising CD56+ cells as the dominant cell type, said cell type being dominant when the proportion of said cell type in said population exceeds 50%.
EP01943584A 2000-06-07 2001-06-07 Method for obtaining characterised muscle-derived cell populations and uses Expired - Lifetime EP1292671B9 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CY20101100077T CY1109745T1 (en) 2000-06-07 2010-01-26 METHOD OF TAKING CHARACTERIZED CELLULAR POPULATION FROM MUSCLES AND USES

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
FR0007304A FR2810045B1 (en) 2000-06-07 2000-06-07 METHOD FOR OBTAINING CELLULAR CELLULAR POPULATIONS OF MUSCLE ORIGIN AND USES THEREOF
FR0007304 2000-06-07
PCT/FR2001/001768 WO2001094555A1 (en) 2000-06-07 2001-06-07 Method for obtaining characterised muscle-derived cell populations and uses

Publications (3)

Publication Number Publication Date
EP1292671A1 EP1292671A1 (en) 2003-03-19
EP1292671B1 true EP1292671B1 (en) 2009-11-18
EP1292671B9 EP1292671B9 (en) 2010-05-19

Family

ID=8851058

Family Applications (1)

Application Number Title Priority Date Filing Date
EP01943584A Expired - Lifetime EP1292671B9 (en) 2000-06-07 2001-06-07 Method for obtaining characterised muscle-derived cell populations and uses

Country Status (14)

Country Link
US (3) US20040043008A1 (en)
EP (1) EP1292671B9 (en)
JP (1) JP2003535586A (en)
AT (1) ATE449163T1 (en)
AU (2) AU6612701A (en)
BR (1) BR0111497A (en)
CA (1) CA2411762A1 (en)
CY (1) CY1109745T1 (en)
DE (1) DE60140536D1 (en)
DK (1) DK1292671T3 (en)
ES (1) ES2335747T3 (en)
FR (1) FR2810045B1 (en)
PT (1) PT1292671E (en)
WO (1) WO2001094555A1 (en)

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PT1272204E (en) * 2000-04-14 2007-10-02 Univ Pittsburgh Soft tissue and bone augmentation and bulking utilizing muscle-derived progenitor cells, compositions and treatments thereof
FR2810045B1 (en) * 2000-06-07 2004-09-03 Assist Publ Hopitaux De Paris METHOD FOR OBTAINING CELLULAR CELLULAR POPULATIONS OF MUSCLE ORIGIN AND USES THEREOF
MXPA04011043A (en) * 2002-05-10 2005-02-14 Isolagen Technologies Inc Treatments with autologous fibroblast.
US9617516B2 (en) * 2003-04-25 2017-04-11 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Muscle-derived cells (MDCs) for promoting and enhancing nerve repair and regeneration
WO2006006692A1 (en) * 2004-07-08 2006-01-19 Japan Science And Technology Agency Animal tissue-eccentrically located pluripotent cell proliferating selectively in low-serium medium
FR2890977A1 (en) * 2005-09-19 2007-03-23 Assist Publ Hopitaux De Paris METHOD FOR OBTAINING HUMAN SMOOTH MUSCLE CELLS AND THEIR APPLICATIONS
FR2909559B1 (en) * 2006-12-06 2012-09-07 Inst Vaisseaux Et Du Sang CELLULAR PREPARATIONS FOR USE AS AGENT STIMULATING REVASCULARIZATION
AU2008263134B2 (en) * 2007-05-29 2014-06-19 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Bone augmentation utilizing muscle-derived progenitor compositions, and treatments thereof
EP2206774A1 (en) 2008-12-23 2010-07-14 Genzyme Corporation Muscle-derived cells having differentiation capacities
CN102712897B (en) * 2009-07-09 2018-07-10 詹森生物科技公司 Heart tissue derived cell
FR3074018B1 (en) 2017-11-27 2020-09-11 Lab Francais Du Fractionnement THERAPEUTIC-AIMED CELL CRYOPRESERVATION PROCESS
FR3088541B1 (en) 2018-11-15 2021-11-05 Lab Francais Du Fractionnement CELL CONSERVATION PROCESS FOR THERAPEUTIC AIMS
WO2020243543A1 (en) * 2019-05-31 2020-12-03 Figene, Llc Fibroblast therapy for treatment of duchenne muscular dystrophy
EP4026587A4 (en) * 2019-09-30 2022-11-16 TERUMO Kabushiki Kaisha Method for increasing proportion of cd56+ cells

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5750397A (en) * 1990-03-30 1998-05-12 Systemix, Inc. Human hematopoietic stem cell

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5143842A (en) * 1988-11-01 1992-09-01 The University Of Colorado Foundation, Inc. Media for normal human muscle satellite cells
US5538722A (en) * 1989-06-13 1996-07-23 Stanford University Isolation, growth, differentiation and genetic engineering of human muscle cells
US5574205A (en) * 1989-07-25 1996-11-12 Cell Genesys Homologous recombination for universal donor cells and chimeric mammalian hosts
DK0412700T3 (en) * 1989-08-03 1998-04-06 Univ California Method for Isolating Fetal Cytrophoblast Cells
US5196315A (en) * 1990-05-01 1993-03-23 The Johns Hopkins University Human neuronal cell line
AU2593192A (en) * 1992-09-14 1994-04-12 Oystein Fodstad Detection of specific target cells in specialized or mixed cell population and solutions containing mixed cell populations
DE69430846T2 (en) * 1993-08-27 2003-01-16 Dana Farber Cancer Inst Inc FOR NATURAL KILLER CELLS SPECIFIC ANTIGENS AND IDENTIFYING ANTIBODIES
US6110459A (en) * 1997-05-28 2000-08-29 Mickle; Donald A. G. Transplants for myocardial scars and methods and cellular preparations
CA2324350A1 (en) * 1998-03-23 1999-09-30 Zymogenetics, Inc. Cardiac-derived stem cells
ATE369141T1 (en) * 1998-05-01 2007-08-15 Univ Pittsburgh CELL-MEDIATED GENE ADMINISTRATION BY USING MUSCLE-DERIVATED CELLS TO TREAT THE BONE-RELATED INJURY OR DISORDER
US7368420B1 (en) * 1998-10-02 2008-05-06 Caritas St. Elizabeth's Medical Center Of Boston, Inc. Akt compositions for enhancing survival of cells
CA2378643A1 (en) * 1999-07-23 2001-02-01 Diacrin, Inc. Muscle cells and their use in cardiac repair
US20030113301A1 (en) * 1999-07-23 2003-06-19 Albert Edge Muscle cells and their use in cardiac repair
FR2810045B1 (en) * 2000-06-07 2004-09-03 Assist Publ Hopitaux De Paris METHOD FOR OBTAINING CELLULAR CELLULAR POPULATIONS OF MUSCLE ORIGIN AND USES THEREOF

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5750397A (en) * 1990-03-30 1998-05-12 Systemix, Inc. Human hematopoietic stem cell

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
KUÇI ET AL: "Phenotypic and functional characterization of mobilized peripheral blood CD34+ cells coexpressing different levels of c-Kit", LEUKEMIA RESEARCH, vol. 22, 1998, pages 355 - 363 *
TAYLOR ET AL: "Delivery of primary autologous skeletal myoblasts into the rabbit heart by coronary infusion: a potential approach to myocardial repair", PROCEEDINGS OF THE ASSOCIATION OF AMERICAN PHYSICIANS, vol. 109, no. 3, 1997, pages 245 - 253, XP008106792 *
TAYLOR ET AL: "Regenerating functional myocardium: improved performance after skeletal myoblast transplantation", NATURE MEDICINE, vol. 4, no. 8, August 1998 (1998-08-01), pages 929 - 933 *

Also Published As

Publication number Publication date
CY1109745T1 (en) 2014-09-10
AU6612701A (en) 2001-12-17
US20060088508A1 (en) 2006-04-27
CA2411762A1 (en) 2001-12-13
EP1292671A1 (en) 2003-03-19
ES2335747T3 (en) 2010-04-05
PT1292671E (en) 2010-02-05
BR0111497A (en) 2004-04-06
FR2810045B1 (en) 2004-09-03
EP1292671B9 (en) 2010-05-19
ATE449163T1 (en) 2009-12-15
AU2001266127B2 (en) 2007-01-04
DE60140536D1 (en) 2009-12-31
US20110014159A1 (en) 2011-01-20
US20040043008A1 (en) 2004-03-04
JP2003535586A (en) 2003-12-02
WO2001094555A1 (en) 2001-12-13
DK1292671T3 (en) 2010-03-15
FR2810045A1 (en) 2001-12-14

Similar Documents

Publication Publication Date Title
JP6755850B2 (en) Use of mesenchymal stem cells
EP1984490B1 (en) Method for culturing cells derived from the adipose tissue and uses thereof
EP1349919B1 (en) Extramedullary adipose tissue cells and use thereof for regenerating cardiac tissue
US20110014159A1 (en) Method for obtaining characterized muscle-derived cell populations and uses
CN101437938B (en) Cardiac stem cells
US8900573B2 (en) Immune privileged and modulatory progenitor cells
JP5570814B2 (en) Muscle-derived cells for the treatment of gastroesophageal pathology and their preparation and use
US11963983B2 (en) Methods of cardiac repair
EP3636271A1 (en) Therapeutic agent for dilated cardiomyopathy
EP2092057B1 (en) Use of bone marrow cells for long term culture of pancreatic islet cells
Guo et al. Myoblast-based cardiac repair: xenomyoblast versus allomyoblast transplantation
JP7117704B1 (en) Preventive and/or therapeutic agent for diabetes
US20230149523A1 (en) Treatment of autoimmunity and transplant rejection through establishment and/or promotion of tolerogenic processes by fibroblast-mediated reprogramming of antigen presenting cells
WO2022176735A1 (en) Prophylactic and/or therapeutic agent for diabetes
AU2014218470A1 (en) Use of bone marrow cells for long term culture of pancreatic islet cells.

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20030106

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

RIN1 Information on inventor provided before grant (corrected)

Inventor name: VILQUIN, JEAN-THOMAS

Inventor name: MAROLLEAU, JEAN-PIERRE

Inventor name: TERNAUX, BRIGITTE

Inventor name: TREMBLAY, JACQUES

Inventor name: ROBERT, ISABELLE

17Q First examination report despatched

Effective date: 20060710

17Q First examination report despatched

Effective date: 20060710

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

Free format text: NOT ENGLISH

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REF Corresponds to:

Ref document number: 60140536

Country of ref document: DE

Date of ref document: 20091231

Kind code of ref document: P

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: BRAUNPAT BRAUN EDER AG

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20100129

REG Reference to a national code

Ref country code: GR

Ref legal event code: EP

Ref document number: 20100400202

Country of ref document: GR

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2335747

Country of ref document: ES

Kind code of ref document: T3

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20100819

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20120622

Year of fee payment: 12

Ref country code: TR

Payment date: 20120601

Year of fee payment: 12

Ref country code: NL

Payment date: 20120523

Year of fee payment: 12

Ref country code: LU

Payment date: 20120525

Year of fee payment: 12

Ref country code: DK

Payment date: 20120522

Year of fee payment: 12

Ref country code: IE

Payment date: 20120528

Year of fee payment: 12

Ref country code: CH

Payment date: 20120626

Year of fee payment: 12

Ref country code: MC

Payment date: 20120521

Year of fee payment: 12

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GR

Payment date: 20120524

Year of fee payment: 12

Ref country code: FI

Payment date: 20120523

Year of fee payment: 12

Ref country code: SE

Payment date: 20120627

Year of fee payment: 12

Ref country code: GB

Payment date: 20120625

Year of fee payment: 12

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20120626

Year of fee payment: 12

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: BE

Payment date: 20120628

Year of fee payment: 12

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20120625

Year of fee payment: 12

Ref country code: FR

Payment date: 20120716

Year of fee payment: 12

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: PT

Payment date: 20120521

Year of fee payment: 12

Ref country code: CY

Payment date: 20120521

Year of fee payment: 12

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: AT

Payment date: 20120523

Year of fee payment: 12

REG Reference to a national code

Ref country code: PT

Ref legal event code: MM4A

Free format text: LAPSE DUE TO NON-PAYMENT OF FEES

Effective date: 20131209

BERE Be: lapsed

Owner name: INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE

Effective date: 20130630

Owner name: ASSISTANCE PUBLIQUE, HOPITAUX DE PARIS

Effective date: 20130630

Owner name: ASSOCIATION FRANCAISE CONTRE LES MYOPATHIES

Effective date: 20130630

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 60140536

Country of ref document: DE

REG Reference to a national code

Ref country code: NL

Ref legal event code: V1

Effective date: 20140101

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130608

Ref country code: CY

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130415

Ref country code: PT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20131209

Ref country code: MC

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130701

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: SE

Ref legal event code: EUG

REG Reference to a national code

Ref country code: DK

Ref legal event code: EBP

Effective date: 20130630

REG Reference to a national code

Ref country code: AT

Ref legal event code: MM01

Ref document number: 449163

Country of ref document: AT

Kind code of ref document: T

Effective date: 20130607

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20130607

REG Reference to a national code

Ref country code: GR

Ref legal event code: ML

Ref document number: 20100400202

Country of ref document: GR

Effective date: 20140103

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130607

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

REG Reference to a national code

Ref country code: FR

Ref legal event code: ST

Effective date: 20140228

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130630

Ref country code: CY

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130607

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130607

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130630

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130630

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130607

Ref country code: NL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140101

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140101

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130701

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130607

Ref country code: AT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130607

Ref country code: GR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140103

REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Ref document number: 60140536

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: C12N0005080000

Ipc: C12N0005073000

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20140708

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 60140536

Country of ref document: DE

Effective date: 20140101

Ref country code: DE

Ref legal event code: R079

Ref document number: 60140536

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: C12N0005080000

Ipc: C12N0005073000

Effective date: 20140602

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130630

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130608

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130607

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130607